US20160095815A1 - Substance for restoring normal co-expression and interaction between the lox and nrage proteins - Google Patents

Substance for restoring normal co-expression and interaction between the lox and nrage proteins Download PDF

Info

Publication number
US20160095815A1
US20160095815A1 US14/969,783 US201514969783A US2016095815A1 US 20160095815 A1 US20160095815 A1 US 20160095815A1 US 201514969783 A US201514969783 A US 201514969783A US 2016095815 A1 US2016095815 A1 US 2016095815A1
Authority
US
United States
Prior art keywords
lox
nrage
expression
extract
skin
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US14/969,783
Inventor
Charbel Bouez
Claudine Gleyzal
Isabelle Orly
Valerie Andre
Pascal Sommer
Corinne Reymermier
Odile Damour
Eric Perrier
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Centre National de la Recherche Scientifique CNRS
Universite Claude Bernard Lyon 1 UCBL
BASF Beauty Care Solutions France SAS
Original Assignee
Centre National de la Recherche Scientifique CNRS
Universite Claude Bernard Lyon 1 UCBL
BASF Beauty Care Solutions France SAS
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 Centre National de la Recherche Scientifique CNRS, Universite Claude Bernard Lyon 1 UCBL, BASF Beauty Care Solutions France SAS filed Critical Centre National de la Recherche Scientifique CNRS
Priority to US14/969,783 priority Critical patent/US20160095815A1/en
Publication of US20160095815A1 publication Critical patent/US20160095815A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K8/00Cosmetics or similar toiletry preparations
    • A61K8/18Cosmetics or similar toiletry preparations characterised by the composition
    • A61K8/96Cosmetics or similar toiletry preparations characterised by the composition containing materials, or derivatives thereof of undetermined constitution
    • A61K8/97Cosmetics or similar toiletry preparations characterised by the composition containing materials, or derivatives thereof of undetermined constitution from algae, fungi, lichens or plants; from derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/168Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from plants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/17Gnetophyta, e.g. Ephedraceae (Mormon-tea family)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/18Magnoliophyta (angiosperms)
    • A61K36/185Magnoliopsida (dicotyledons)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/18Magnoliophyta (angiosperms)
    • A61K36/185Magnoliopsida (dicotyledons)
    • A61K36/45Ericaceae or Vacciniaceae (Heath or Blueberry family), e.g. blueberry, cranberry or bilberry
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/18Magnoliophyta (angiosperms)
    • A61K36/185Magnoliopsida (dicotyledons)
    • A61K36/48Fabaceae or Leguminosae (Pea or Legume family); Caesalpiniaceae; Mimosaceae; Papilionaceae
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/18Magnoliophyta (angiosperms)
    • A61K36/185Magnoliopsida (dicotyledons)
    • A61K36/54Lauraceae (Laurel family), e.g. cinnamon or sassafras
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/18Magnoliophyta (angiosperms)
    • A61K36/185Magnoliopsida (dicotyledons)
    • A61K36/77Sapindaceae (Soapberry family), e.g. lychee or soapberry
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K8/00Cosmetics or similar toiletry preparations
    • A61K8/18Cosmetics or similar toiletry preparations characterised by the composition
    • A61K8/96Cosmetics or similar toiletry preparations characterised by the composition containing materials, or derivatives thereof of undetermined constitution
    • A61K8/97Cosmetics or similar toiletry preparations characterised by the composition containing materials, or derivatives thereof of undetermined constitution from algae, fungi, lichens or plants; from derivatives thereof
    • A61K8/9778Gnetophyta, e.g. Ephedraceae [Mormon-tea family]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K8/00Cosmetics or similar toiletry preparations
    • A61K8/18Cosmetics or similar toiletry preparations characterised by the composition
    • A61K8/96Cosmetics or similar toiletry preparations characterised by the composition containing materials, or derivatives thereof of undetermined constitution
    • A61K8/97Cosmetics or similar toiletry preparations characterised by the composition containing materials, or derivatives thereof of undetermined constitution from algae, fungi, lichens or plants; from derivatives thereof
    • A61K8/9783Angiosperms [Magnoliophyta]
    • A61K8/9789Magnoliopsida [dicotyledons]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0014Skin, i.e. galenical aspects of topical compositions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/04Antipruritics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P39/00General protective or antinoxious agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61QSPECIFIC USE OF COSMETICS OR SIMILAR TOILETRY PREPARATIONS
    • A61Q19/00Preparations for care of the skin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61QSPECIFIC USE OF COSMETICS OR SIMILAR TOILETRY PREPARATIONS
    • A61Q19/00Preparations for care of the skin
    • A61Q19/004Aftersun preparations
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61QSPECIFIC USE OF COSMETICS OR SIMILAR TOILETRY PREPARATIONS
    • A61Q19/00Preparations for care of the skin
    • A61Q19/08Anti-ageing preparations
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5011Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing antineoplastic activity
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/573Immunoassay; Biospecific binding assay; Materials therefor for enzymes or isoenzymes
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2800/00Properties of cosmetic compositions or active ingredients thereof or formulation aids used therein and process related aspects
    • A61K2800/40Chemical, physico-chemical or functional or structural properties of particular ingredients
    • A61K2800/59Mixtures
    • A61K2800/596Mixtures of surface active compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2800/00Properties of cosmetic compositions or active ingredients thereof or formulation aids used therein and process related aspects
    • A61K2800/70Biological properties of the composition as a whole

Definitions

  • homeostasis is maintained by a balance between cell proliferation, cell differentiation towards a defined function, and programmed or unprogrammed cell death.
  • the skin for example, can be considered as an organ of which one specific function is to protect the organism from external aggressions. Impermeability to the outside is assured by a highly keratinized corneal layer composed of dead cells.
  • the rhythm of the life of epidermal cells, or keratinocytes is therefore governed by a succession of changes leading to this differentiation into keratinized and dead cells: (1) maintenance of initiator cells (“stem” cells of the basal cell layer of the epidermis), (2) asymmetric division (each stem cell giving two sister cells), (3) differentiation (formation of the suprabasal cell layers of the epidermis from one of the “initiator sister” cells), (4) acquisition of a resistance to apoptotic cell death, (5) change into cornified cells, and (6) programmed cell death of the upper cell layers of the epidermis. Any modification of this balance can be pathological.
  • LOX belongs to the family of the lysyl oxidases (LOs), which are amine oxidases dependent on copper. 5 LO genes have been characterized to date: LOX LOXL, LOXL2, LOXL3 and LOXL4. Where LOX is known for its role in the crosslinking of collagen ex vivo and in vivo, LOXL is clearly associated with the homeostasis of elastic fibers, The in vivo role of the other isoforms is not known.
  • LOs are synthesized by a variety of cells such as fibroblasts, smooth muscle cells, endothelial cells and keratinocytes. This enzyme is therefore present in numerous tissues such as the skin, the liver, the kidneys, the spleen and the aorta, at both extracellular and intracellular levels.
  • LOX extracellular matrix
  • ECM extracellular matrix
  • ⁇ -aminopropionitrile binds to the active site of LOX in competition with alkylamine substrates; LOs use ⁇ -APN as substrate, thereby forming a Schiff base but without releasing the aldehyde product, which blocks the active site covalently without having an effect on synthesis.
  • said document relates to lysyl oxidases as a whole and not specifically to the LOX isoform; furthermore, it relates exclusively to inhibition of the dedifferentiation of cells cultivated in vitro, providing no teaching on the role of lysyl oxidases, or a fortiori of LOX, in maintenance of the balance between proliferation, differentiation and apoptosis, nor does it provide any teaching on novel partners or substrates for lysyl oxidases or LOX.
  • LOX has been located in the epidermis, its expression being regulated as a function of the level of differentiation of keratinocytes (Noblesse et al., Lysyl oxidase-like and lysyl oxidase are present in the dermis and epidermis of a skin equivalent and in human skin and are associated to elastic fibers, J. Invest. Dermatol., 2004, 122, 621-630).
  • the studies carried out hitherto have not made it possible to define the role of LOX in keratinocytes or to search for its partners in these cells, which make little or no collagen or elastin.
  • the LOX gene is clearly associated with maintenance of the non-tumoral phenotype of cells.
  • the LOX enzyme is a suppresser of the ras oncogene and that somatic mutations in the gene are associated with various cancers (Contente et al., Expression of gene rrg is associated with reversion of NIH 3T3 transformed by LTR-c-H-ras, Science, 1990, 249, 796-798; Csiszar et al., Somatic mutations of the lysyl oxidase gene on chromosome 5q23.1 in colorectal tumors, Int. J. Cancer, 2002, 97, 636-642).
  • LOX probably acts according to the availability and regulation of its cellular substrates and partners, but the latter are still unknown to those skilled in the art.
  • apoptosis is used to describe a particular form of cell death whose morphological characteristics differ from those of necrosis.
  • Apoptosis is a process of programmed cell death that requires the acquisition of caspases.
  • the cells acquire particularly remarkable morphological characteristics, such as condensation of the chromatin and fragmentation of the nucleus, leading to their self-destruction and their elimination from the tissue without damaging the adjacent cells.
  • Apoptosis corresponds to the natural death of cells in the course of their development or during homeostasis.
  • the prior art provides information about a number of agents and mechanisms that are involved in regulating the cell cycle via the cellular processes of apoptosis and differentiation,
  • the NRAGE protein comprises 778 amino acids. In the central region it carries a first domain characteristic of the MAGE proteins: the MAGE homology domain (MHD-1), and in the N-terminal region it carries a second domain: MHD-2, present only in certain isoforms. These two domains have zones rich in lysyl residues. Between these two domains there is a region called IRD (interspersed repeat domain), which does not exist in any other protein currently known,
  • the NRAGE protein is involved in controlling apoptosis via different pathways.
  • NRAGE By interacting with p75NTR, which is the “low affinity” receptor of neutrophilic factors (NGF) or TNF, NRAGE can block the progression of the cell cycle and thus is proapoptotic via the caspase pathway.
  • NNF neutrophilic factors
  • NRAGE is also proapoptotic by interacting with cytoplasmic inhibitors of apoptotic proteins, IAP.
  • NRAGE can also act directly on the activity of nuclear homeo factors, such as the factors Msx and Dix, which take part in the morphogenic regulation of tissues.
  • NRAGE Although the expression of NRAGE is ubiquitous, the prior art does not refer to its presence in the skin, nor does it provide any data giving information about a possible relationship between LOX and apoptosis or about a link between LOX and NRAGE.
  • the prior art does not provide the identity of novel partners or substrates for LOX, especially those taking part in maintenance of the cellular balance between proliferation, differentiation and apoptosis; moreover, the prior art does not provide the identity of these novel partners or substrates either in epithelial cells (particularly keratinocytes) or in any other cell type.
  • the prior art gives no information about the possible variations in the expression of LOX in the epidermis (especially LOX expressed by keratinocytes) as a function of age or of the existence or non-existence of exposure to UV or other types of aggression, or in the case of diseases affecting the skin (psoriasis, graft-versus-host reaction, cancers, etc.).
  • the prior art does not provide information about the possible presence of NRAGE in the skin, whether it be healthy skin, skin altered by age or UV or subjected to other types of aggression, or diseased skin.
  • the prior art does not offer known models for studying NRAGE in cells of epithelial origin or in the epidermis.
  • the prior art does not provide a model for identifying active principles capable of modulating the expression of LOX and/or NRAGE in keratinocytes.
  • LOX is expressed in the epidermis, its expression being regulated as a function of the level of differentiation of the keratinocytes (Noblesse et al., 2004).
  • the prior art therefore fails to provide active principles capable of modulating the expression of intracellular partners for LOX (such as NRAGE), which may or may not be associated with modulation of the expression of LOX for the purpose of acting on cell regulation. In this context it is also very difficult to obtain objective criteria for judging the impact of these active principles.
  • LOX such as NRAGE
  • the main object of the invention is to solve the technical problems referred to above, especially the technical problem relating to the provision of a method of identifying active principles for improving the interaction between the LOX and NRAGE proteins in cases where the interaction between LOX and NRAGE is absent or altered.
  • the aim of the invention is to regulate the cellular balance between proliferation, differentiation and apoptosis in cases where it is disturbed, as in the case of skin altered by age or UV or subjected to other types of aggression, and/or in the case of pathological situations such as psoriasis, eczema, graft-versus-host reaction, lichen planus and/or cancerous diseases.
  • the invention further relates to the use of an active principle that modulates the expression of LOX and/or NRAGE for regulating the cellular balance between proliferation, differentiation and apoptosis, particularly in cases where it is disturbed, as in the case of skin altered by age or UV or subjected to other types of aggression, and/or in the case of pathological situations such as psoriasis, eczema, graft-versus-host reaction, and cancerous scars and/or diseases.
  • the invention relates to active principles for improving the interaction between the LOX and NRAGE proteins in cases where the interaction between LOX and NRAGE is absent or altered.
  • the invention makes it possible to solve the technical problem that consists in providing a method of locating the expression of NRAGE and following this expression, especially on a cellular model of skin.
  • the invention makes it possible to solve the technical problem that consists in providing a method of diagnosing a state associated with poor regulation of the interaction between LOX and NRAGE.
  • LOX the isoform of the human protein lysyl oxidase, LOX, as defined in particular by the amino acid sequence SEQ ID no. 1;
  • NRAGE the human protein NRAGE as defined in particular by the amino acid sequence SEQ ID no. 2;
  • modulation of the expression of LOX modulation of the gene coding for LOX, especially modulation of the expression of the messenger RNA coding for LOX, but also modulation of the synthesis of LOX from this messenger RNA, as well as modulation of the activity of LOX;
  • modulation of the expression of NRAGE modulation of the gene coding for NRAGE, especially modulation of the expression of the messenger RNA coding for NRAGE, but also modulation of the synthesis of NRAGE from this messenger RNA, as well as modulation of the biological effect of NRAGE.
  • Active principles which are considered to be effective on LOX are preferably those affording a difference of about ⁇ 50% in the expression of the mRNA of LOX and/or a difference of about ⁇ 15% in the expression of LOX and/or the activity of LOX on a model, comprising at least one cell type exhibiting LOX expression and/or activity, in contact with these active principles, compared with the level of LOX expression and/or activity in a control model (generally without being brought into contact with the active principles).
  • Active principles which are considered to be effective on NRAGE are preferably those affording a difference of about ⁇ 50% in the expression of the mRNA of NRAGE and/or a difference of about ⁇ 15% in the expression of NRAGE and/or the biological effect of NRAGE on a model, comprising at least one cell type exhibiting NRAGE expression and/or activity, in contact with these active principles, compared with the level of NRAGE expression and/or activity in a control model (generally without being brought into contact with the active principles).
  • the present invention relates to the use of an effective amount of at least one substance that modulates the expression and/or activity of at least the LOX protein of sequence ID no. 1, and/or that modulates the expression and/or activity of at least the NRAGE protein of sequence ID no. 2, for the manufacture of a composition for modulating the interaction between LOX and NRAGE in order to regulate the cellular balance between proliferation, differentiation and apoptosis, particularly in cases where the balance between these phenomena is disturbed, and especially in cases where the interaction between LOX and NRAGE is absent or altered.
  • the invention relates to the use of an effective amount of at least one substance that modulates the expression and/or activity of LOX of sequence ID no. 1, and/or that modulates the expression and/or activity of NRAGE of sequence ID no. 2, for the manufacture of a composition for improving the interaction between the LOX and NRAGE proteins in cases where the interaction between LOX and NRAGE is absent or altered.
  • the invention further relates to the use of an effective amount of at least one substance that modulates the expression and/or activity of at least the NRAGE protein of sequence ID no. 2, and that optionally modulates the expression and/or activity of the LOX protein of sequence ID no. 1, for the manufacture of a composition for preventing or treating at least one state in which the cellular balance between proliferation, differentiation and apoptosis is absent or altered.
  • the expression of LOX and/or NRAGE is modulated in epithelial cells, particularly in keratinocytes.
  • the purpose of said substance is to modulate the interaction between the LOX protein and the NRAGE protein, said interaction occurring especially in the IRD domain of the NRAGE protein.
  • the interaction between LOX and NRAGE comprises the polymerization of NRAGE, particularly its dimerization, by LOX-induced enzymatic catalysis.
  • the interaction between LOX and NRAGE involves the production of H 2 O 2 resulting from the catalytic activity of LOX, the H 2 O 2 then activating other molecules, for example neutral sphingomyelinase or NF-kB.
  • the interaction between LOX and NRAGE involves a non-enzymatic activity of LOX, particularly an activity exerted by the pro-region (A22-D169) of LOX.
  • the purpose of the substance is to treat and/or prevent a condition selected from the group consisting of exposure of cells to a stress, particularly exposure of cells to heat, or exposure of cells to radiation, particularly solar radiation, or exposure of cells to a toxic agent, for example a chemical or microbiological agent, skin ageing, lichen planus, graft-versus-host reaction (GVH), eczema, psoriasis and a cancer, particularly an epithelial cancer.
  • a condition selected from the group consisting of exposure of cells to a stress, particularly exposure of cells to heat, or exposure of cells to radiation, particularly solar radiation, or exposure of cells to a toxic agent, for example a chemical or microbiological agent, skin ageing, lichen planus, graft-versus-host reaction (GVH), eczema, psoriasis and a cancer, particularly an epithelial cancer.
  • a condition selected from the group consisting of exposure of cells to a stress, particularly exposure of cells to heat, or exposure of
  • the purpose of the substance is to reduce cell proliferation in the case of hyperproliferation, particularly in a cancer, more particularly an epithelial cancer and most particularly a cutaneous epithelial cancer, of basocellular or spinocellular type, psoriasis or eczema.
  • the purpose of the substance is to reduce apoptosis in the epidermis in the case of substantial apoptosis, particularly during skin ageing, exposure of the skin to a stress, particularly exposure to heat, or exposure of the skin to radiation, particularly solar radiation, or exposure of the skin to a toxic agent, for example a chemical or microbiological agent, or graft-versus-host reaction (GVH).
  • a toxic agent for example a chemical or microbiological agent, or graft-versus-host reaction (GVH).
  • the purpose of the substance is to increase cell proliferation in the case of cell hypoproliferation in the epidermis, particularly during ageing, exposure of the skin to heat, exposure of the skin to radiation, particularly solar radiation, or graft-versus-host reaction (GVH).
  • VH graft-versus-host reaction
  • the purpose of the substance is to stimulate the expression of LOX, and optionally to inhibit the expression of NRAGE, during skin ageing, exposure of the skin to a stress, particularly exposure to heat, or exposure of the skin to radiation, particularly solar radiation, or exposure of the skin to a toxic agent, for example a chemical or microbiological agent, or graft-versus-host reaction (GVH).
  • a toxic agent for example a chemical or microbiological agent, or graft-versus-host reaction (GVH).
  • the purpose of the substance is to stimulate the expression and/or activity of NRAGE, and optionally to inhibit the expression and/or activity of LOX, in the epidermis in order to prevent or treat psoriasis or eczema.
  • the purpose of the substance is to stimulate the expression of LOX and NRAGE in order to prevent or treat a cancer, particularly an epithelial cancer and more particularly a cutaneous epithelial cancer, of basocellular or spinocellular type, or lichen planus.
  • said composition is a cosmetic, neutraceutical, dermo-pharmaceutical or pharmaceutical composition.
  • the cellular balance between proliferation, differentiation and apoptosis is the balance between proliferation, differentiation and apoptosis of the keratinocytes.
  • the starting material used to prepare the active principles in the case of plants (preferably roots, stems, barks, flowers, fruits, seeds, germs, gums, exudates, leaves or whole plants) or proteins, may or may not be sterilized by radiation, for example beta or gamma radiation preferably at a dose of 5 kGy, and is then reduced to powder if necessary, for example by grinding at room temperature. The powder is then e.g.
  • a polar solvent for example water, an alcohol, a glycol such as butylene glycol, or a polyol, and/or a mixture of polar solvents, advantageously a mixture of water with an alcohol, glycol or polyol (such as ethanol, glycerol, butylene glycol and other glycols, xylitol, etc.) in variable proportions, preferably a 75/25 or 50/50 water/butylene glycol mixture, or in an apolar solvent, for example an alkane, or a mixture of apolar solvents, or in a mixture of polar and apolar solvents.
  • a polar solvent for example water, an alcohol, a glycol such as butylene glycol, or a polyol, and/or a mixture of polar solvents, advantageously a mixture of water with an alcohol, glycol or polyol (such as ethanol, glycerol, butylene glycol and other glycols, xylitol, etc.)
  • the sample is preferably clarified by decantation or centrifugation and then filtered, preferably on a 0.45 ⁇ m or 0.22 ⁇ m filter.
  • the starting material used to prepare the active principles in the case of characterized molecules (e.g. molecules obtained by synthesis or hemisynthesis, biological molecules obtained by purification), is diluted in a solvent, preferably water or dimethyl sulfoxide (in a concentration preferably of between 10 ⁇ 6 M and 10 ⁇ 2 M and particularly preferably in the order of 10 ⁇ 4 M, or preferably of between 1% weight/weight and 5% weight/weight, depending on the molecules).
  • a solvent preferably water or dimethyl sulfoxide (in a concentration preferably of between 10 ⁇ 6 M and 10 ⁇ 2 M and particularly preferably in the order of 10 ⁇ 4 M, or preferably of between 1% weight/weight and 5% weight/weight, depending on the molecules).
  • a solvent preferably water or dimethyl sulfoxide (in a concentration preferably of between 10 ⁇ 6 M and 10 ⁇ 2 M and particularly preferably in the order of 10 ⁇ 4 M, or preferably of between 1% weight/weight and 5% weight/weight, depending
  • the active principles obtained by one of the methods described above are used in a final concentration preferably of between 0.01% volume/volume (v/v) and 10% (v/v) and particularly preferably of between 0.1% and 1% (v/v).
  • said substance is selected from the group consisting of a soya extract, an ephedra extract, a hop extract and a cinnamon extract.
  • the invention relates to a method of identifying at least one active principle described above, the purpose of which is in particular to modulate the interaction between LOX and NRAGE in order to prevent or treat at least one state in which the cellular balance between proliferation, differentiation and apoptosis is absent or altered, characterized in that it comprises:
  • analyzing the expression of LOX and/or NRAGE especially to identify an active principle that modulates the expression and/or activity of LOX and/or NRAGE in order to improve the cellular balance between proliferation, differentiation and apoptosis.
  • the invention further relates to a method of identifying at least one active principle for modulating the interaction between LOX and NRAGE in order to improve the interaction between the LOX and NRAGE proteins in cases where the interaction between LOX and NRAGE is absent or altered, characterized in that it comprises:
  • analyzing the expression of LOX and/or NRAGE especially to identify an active principle that modulates the expression and/or activity of LOX and/or NRAGE in order to improve the interaction between the LOX and NRAGE proteins in cases where the interaction between LOX and NRAGE is absent or altered.
  • the balance between the proliferation, differentiation and apoptosis of said living cells capable of expressing the LOX and/or NRAGE protein, or the interaction between the LOX and NRAGE proteins, is absent or altered before they are brought into contact with the active principle.
  • the living cells are epithelial cells, particularly keratinocytes.
  • the method comprises analyzing the expression of the messenger RNAs of LOX and/or NRAGE.
  • the method comprises using quantitative RT-PCR with the following primers in particular:
  • the method comprises analyzing the kinetics of expression of the messenger RNAs, for example by quantitative RT-PCR.
  • the invention further relates to a method for the cosmetic care or therapeutic treatment of a subject in whom the balance between the cellular phenomena of proliferation, differentiation and apoptosis in at least one cell type is disturbed, said method comprising improving the interaction between LOX and NRAGE by the application or administration of a substance that modulates the expression of LOX and/or NRAGE.
  • the invention further relates to a method for the cosmetic care or therapeutic treatment of a subject in whom the interaction between LOX and NRAGE is absent or altered, said method comprising the application or administration of a substance that modulates the expression and/or activity of LOX of sequence ID no. 1, and/or that modulates the expression and/or activity of NRAGE of sequence ID no. 2.
  • the invention further relates to a method for the cosmetic care or therapeutic treatment of a subject presenting with a state in which the cellular balance between proliferation, differentiation and apoptosis is absent or altered, said method comprising the application or administration of a substance that modulates the expression and/or activity of NRAGE, and optionally that modulates the expression and/or activity of LOX.
  • the invention relates to a process for the preparation of a composition, comprising:
  • the mixing of the active principle with at least one excipient in order to produce a cosmetic, neutraceutical, dermopharmaceutical or pharmaceutical composition for preventing or treating at least one state in which the cellular balance between proliferation, differentiation and apoptosis is absent or altered.
  • the invention further relates to a process for the preparation of a composition, comprising:
  • the invention relates to a method of locating NRAGE, comprising the use of at least one anti-NRAGE antibody for detecting and locating the presence of NRAGE, particularly in a model of reconstructed skin comprising at least keratinocytes, or a section of skin, preferably a section of skin originating from a person whose epidermis exhibits a state in which the cellular balance between proliferation, differentiation and apoptosis is absent or altered.
  • the invention relates to the use of an anti-NRAGE antibody for the preparation of a composition for detecting a modulation of the expression of NRAGE at the cellular level, particularly in epithelial cells and preferably in keratinocytes.
  • the purpose of the composition is to detect a state in which the cellular balance between proliferation, differentiation and apoptosis, or the interaction between LOX and NRAGE, particularly in the epidermis, is absent or altered, said state being selected from the group consisting of exposure of cells to a stress, particularly exposure of cells to heat, or exposure of cells to radiation, particularly solar radiation, or exposure of cells to a toxic agent, for example a chemical or microbiological agent, skin ageing, lichen planus, graft-versus-host reaction (GVH), eczema, psoriasis and a cancer, particularly an epithelial cancer and more particularly a cutaneous epithelial cancer, of basocellular or spinocellular type.
  • a toxic agent for example a chemical or microbiological agent, skin ageing, lichen planus, graft-versus-host reaction (GVH), eczema, psoriasis and a cancer, particularly an epithelial cancer and more particularly a cutaneous epit
  • the composition also comprises an anti-LOX antibody for detecting a modulation of the expression of LOX at the cellular level, particularly in epithelial cells and preferably in keratinocytes.
  • composition also comprises an anti-LOX antibody for detecting a modulation of the expression of LOX in keratinocytes.
  • the keratinocytes are human keratinocytes.
  • LOX the protein which, being present at the intersection of the proliferation, differentiation and apoptosis pathways, is capable of controlling cellular homeostasis.
  • the inventors have also discovered that the NRAGE protein is present in particular in the epidermis and is expressed in particular by the keratinocytes. This discovery also enables the interaction between LOX and NRAGE to be made an action target for cosmetic care or treatments for restoring cellular homeostasis in situations where it is disturbed.
  • the next step consisted in determining whether the identification of this potential partnership between LOX and NRAGE could correspond to a direct physical interaction between these two proteins. This was carried out in particular by a technique involving co-immunoprecipitation of the LOX and NRAGE proteins produced after transfection of their genes into Cos7 cells. The results obtained on the one hand confirmed the physical interaction between LOX and NRAGE and on the other hand made it possible to specify that LOX interacted with a particular region of NRAGE called IRD (interspersed repeat domain), which is a region specific to NRAGE in the family of the MAGE proteins (cf. Example 3).
  • IRD interspersed repeat domain
  • the inventors identified the expression of NRAGE in both the dermis and the epidermis of the skin (cf. Examples 4 and 5).
  • the inventors used a method of locating the expression of NRAGE in the skin.
  • NRAGE was not expressed homogeneously—as would have been expected of a protein whose presence is generally described as ubiquitous—but in the form of an expression gradient.
  • the epidermis is a stratified epithelium resting on a basal membrane which anchors it to the dermis. Its thickness varies from 60 to 100 ⁇ m on average and it consists of 4 continuous layers, from the deepest to the surface, resulting from a progressive differentiation of the keratinocytes:
  • the basal cell layer (single row of cells)
  • the prickle cell layer (5 to 6 monocellular layers)
  • the horny layer (5 to 10 monocellular layers)
  • NRAGE is absent in the basal cell layer and appears in the first layers of differentiated keratinocytes, the labeling increasing in the horny layers.
  • the labeling of the first suprabasal cell layers of the epidermis corresponds to that observed for LOX, which appears from the basal cell layer upwards.
  • the labeling of LOX decreases in the upper layers of the epidermis, where NRAGE is present, so a distinction is made between the following 3 zones:
  • Observations at the cellular level show that LOX and NRAGE appear at the periphery of the cell, particularly in the submembranous region, NRAGE also appearing in the cytoplasm (cf. Examples 4 and 5).
  • the inventors thus identified, for the first time, the presence of NRAGE in the skin, both in the dermis and the epidermis. They also showed that the epidermis comprises a part in which both the LOX and NRAGE proteins are expressed and share the same location at the cellular level, especially in the keratinocytes, said location being the submembranous peripheral zone (NRAGE also being present in the cytoplasm).
  • the inventors thus demonstrated fulfilment of the conditions for the direct interaction they had identified in vitro to be able to take place in the epidermis in the zone where both LOX and NRAGE are located.
  • the inventors also demonstrated, unexpectedly, that skin ageing and a number of pathological situations are accompanied by disturbances of the expression of LOX and/or NRAGE in the epidermis.
  • the skin of elderly persons is characterized by a hypoproliferative epidermis that is very thin (reduced to a few cell layers) and hyperkeratinized.
  • NRAGE is strongly expressed, is located in the cytoplasm and appears from the first suprabasal cell layer upwards, without an expression gradient.
  • the present invention makes it possible to stimulate the expression (and/or activity) of LOX, with or without inhibiting the expression (and/or activity) of NRAGE, so as to reinduce a regulated differentiation zone and/or apoptosis zone, in particular by restoring a LOX and NRAGE co-expression zone. This also thickens the skin, especially the epidermis.
  • the purpose of the present invention is especially to correct or prevent the effects of skin ageing.
  • GVH is a disease which can arise following allografts of hemopoietic stem cells. It is associated with the effect of the immune cells (lymphocytes) contained in the graft on the patient's normal organs (especially the skin, liver and digestive tract). In the skin it manifests itself in the form of a maculopapular, pruriginous and inflammatory eruption.
  • the present invention makes it possible to stimulate the expression (and/or activity) of LOX, with or without inhibiting the expression (and/or activity) of NRAGE, in order to reinduce a LOX and NRAGE co-expression zone, enabling them to interact and resulting in a reduction in the cutaneous manifestations of GVH, this being accompanied especially by a thickening of the skin, particularly the epidermis.
  • the present invention enables patients suffering from GVH to reduce the cutaneous manifestations of this disease.
  • Lichen is a skin disease of unknown cause which is characterized by the presence of violet, flat, solid, dry and very pruriginous papules a few millimeters in diameter.
  • the inventors identified a very large decrease in the expression of LOX, or even its total absence, and a very irregular expression of NRAGE. These observations certainly reflect a non-existent or very weak level of interaction between the two proteins studied.
  • the present invention makes it possible to stimulate the expression (and/or activity) of LOX, with or without modulating the expression (and/or activity) of NRAGE, in order to reinduce a LOX and NRAGE co-expression zone, enabling them to interact and making it possible to return to a normal epidermal state.
  • the present invention enables patients suffering from lichen planus to treat, reduce or prevent this disease.
  • Psoriasis is a chronic skin disease characterized by erythematosquamous lesions.
  • the fundamental feature of the disease is an increase in the rate of multiplication of the keratinocytes, which is responsible for a more rapid renewal and a thickening of the epidermis.
  • LOX is essentially expressed in the lower part of the epidermis and NRAGE solely in its upper part, the expression of the proteins thus being shifted without there being the overlap zone normally observed in healthy skin.
  • NRAGE is observed only in the cytoplasm and not in the submembranous peripheral zone.
  • inhibition of the expression (and/or activity) of LOX, and/or optionally stimulation, preferably partial stimulation, of the expression (and/or activity) of NRAGE makes it possible in particular to obtain an overlap zone of LOX and NRAGE expression in order to reinduce a regulated proliferation zone that makes it possible to reduce hyperproliferation by promoting apoptosis in the keratinocytes.
  • stimulation of the expression (and/or activity) of NRAGE, and/or inhibition, preferably partial inhibition, of the expression (and/or activity) of LOX makes it possible in particular to obtain an overlap zone of LOX and NRAGE expression in order to reinduce a regulated proliferation zone that makes it possible to reduce hyperproliferation by promoting apoptosis in the keratinocytes.
  • the present invention makes it possible to prevent and/or treat psoriasis or reduce some of its effects.
  • Eczema is a skin complaint characterized clinically by blotches, more or less extensive, localized swellings, and weeping vesicles that subsequently form scabs, accompanied by intense itching. In its chronic phase eczema is complicated by a modification of the skin with thickening. Apoptosis is reduced in the epidermis.
  • NRAGE is weakly expressed and is observed only in the cytoplasm, which certainly reflects a non-existent or very weak level of interaction between the two proteins studied.
  • stimulation of the expression (and/or activity) of NRAGE, and/or inhibition, preferably partial inhibition, of the expression (and/or activity) of LOX makes it possible to increase apoptosis and thereby to reduce some of the effects of eczema.
  • the present invention makes it possible to prevent and/or treat eczema and in particular to reduce some of its effects.
  • epithelial skin carcinoma A distinction is made between two major types of epithelial skin carcinoma:
  • basocellular carcinoma (90% of cases) is an essentially local, slowly developing tumor which virtually never metastasizes. It results from an uncontrolled proliferation of the keratinocytes of the basal cell layer;
  • spinocellular carcinoma (10% of cases) has a much more aggressive local development and can metastasize. It originates from an uncontrolled proliferation of the keratinocytes of the prickle cell layer.
  • the inventors identified the absence of LOX and NRAGE in the invasive cells of the two types of cancer studied (basocellular and spinocellular cancers), with a progressive loss of LOX and NRAGE in the epidermis in the vicinity of the tumors.
  • the inventors also observed that LOX is strongly expressed in the stromal reaction around the tumors, whereas NRAGE is absent in this reaction.
  • stimulation of the expression (and/or activity) of NRAGE and the expression (and/or activity) of LOX makes it possible to restore homeostasis. It is possible to bring about a reversion of the tumoral phenotype, especially in epithelial skin cancers.
  • the inventors provided methods of identifying active principles that modulate the expression of LOX and/or NRAGE in order to restore the control exerted by these proteins via their interaction, for the purpose of identifying active principles for the preparation of compositions, especially cosmetic or pharmaceutical compositions.
  • the expression of LOX is stimulated, with or without inhibiting the expression of NRAGE, in order to reinduce, via modulation of the co-expression of LOX and NRAGE, a regulated differentiation and apoptosis zone that leads to a thickening of the skin.
  • NRAGE In the case of epidermal hyperproliferation (psoriasis, eczema), where NRAGE is underexpressed, the expression of NRAGE is stimulated, with or without (slightly) inhibiting that of LOX, in order to promote apoptosis by reinducing a regulated proliferation zone (LOX/NRAGE overlap) and making it possible to stop the hyperproliferation.
  • apoptosis in the epidermis aged skin, exposure of the skin to a stress, particularly exposure to heat, or exposure of the skin to radiation, particularly solar radiation, or exposure of the skin to a toxic agent, for example a chemical or microbiological agent, or graft-versus-host reaction—GVH)
  • a toxic agent for example a chemical or microbiological agent, or graft-versus-host reaction—GVH
  • the expression of NRAGE is inhibited, with or without stimulating the expression of LOX.
  • the inventors identified the existence of a hitherto unknown link between LOX and apoptosis.
  • the data obtained reflect the antiapoptotic role of LOX in the keratinocytes, said role involving regulation of the proapoptotic protein NRAGE in particular.
  • Active principles were identified in particular by analyzing the expression of the messenger RNAs of LOX and NRAGE, especially on keratinocytes in culture and preferably on human keratinocytes.
  • the active principles whose activity is to be tested are placed in contact with the keratinocytes in culture for a sufficient time and under appropriate conditions for the contact to be effective.
  • the active principles are preferably tested in different concentrations so that any influence of concentration can be detected.
  • the active principles screened in the present invention are of vegetable origin, in particular so as to avoid the problems associated with chemical synthesis.
  • the advantages of active principles of vegetable origin are well known to those skilled in the art, especially in pharmacy, dermopharmacy, neutraceutics and cosmetics.
  • the search for active principles is carried out especially by extracting the total RNAs and then performing quantitative RT-PCR.
  • the preferred primer sequences are those used in Example 13, without implying a limitation.
  • an active principle makes it possible to modulate the expression of the messenger RNA of LOX and/or NRAGE by at least 50% or the expression and/or activity of LOX and/or NRAGE by at least 15%.
  • compositions especially cosmetic, dermopharmaceutical or pharmaceutical compositions.
  • the excipient for these compositions therefore contains e.g. at least one compound selected from the group consisting of preservatives, emollients, emulsifiers, surfactants, moisturizers, thickeners, conditioners, matting agents, stabilizers, antioxidants, texturizing agents, brightening agents, film-forming agents, solubilizers, pigments, colorants, perfumes and sun filters.
  • excipients are preferably selected from the group consisting of amino acids and their derivatives, polyglycerols, esters, cellulose polymers and derivatives, lanolin derivatives, phospholipids, lactoferrins, lactoperoxidases, sucrose-based stabilizers, vitamin E and its derivatives, natural and synthetic waxes, vegetable oils, triglycerides, unsaponifiables, phytosterols, plant esters, silicones and their derivatives, protein hydrolyzates, jojoba oil and its derivatives, liposoluble/water-soluble esters, betaines, amine oxides, plant extracts, sucrose esters, titanium dioxides, glycines and parabens, and particularly preferably from the group consisting of butylene glycol, steareth-2, steareth-21, glycol-15 stearyl ether, cetearyl alcohol, phenoxyethanol, methylparaben, ethylparaben, propylparaben, butylpara
  • compositions are formulated in a form selected from the group consisting of an aqueous or oily solution, a cream or an aqueous or oily gel, particularly in a pot or tube, especially a shower gel or a shampoo; a milk; an emulsion, microemulsion or nanoemulsion, particularly an oil-in-water, water-in-oil, multiple or silicone emulsion; a lotion, particularly in a glass or plastic bottle, a dosing bottle or an aerosol; an ampoule; a syrup; a liquid soap; a hypoallergenic cleansing bar; an ointment; a foam; an injectable solution; an anhydrous, preferably liquid, pasty or solid product, for example in stick form, especially in the form of lipstick; a powder; and a tablet.
  • a form selected from the group consisting of an aqueous or oily solution, a cream or an aqueous or oily gel, particularly in a pot or tube, especially a shower gel or a shampoo;
  • FIG. 1 shows a sequence and a diagram of the NRAGE protein
  • FIG. 2 shows the mean of the results obtained in Example 2 for the two-hybrid interaction
  • FIG. 3 shows the identification of the presence of LOX and NRAGE in reconstructed skin
  • FIG. 4 shows the location of LOX and NRAGE on human skin sections by confocal microscopy
  • FIG. 5 shows the detection of LOX and NRAGE on' human skin sections from a 91-year-old donor
  • FIG. 6 shows the detection of LOX and NRAGE in the skin of a person suffering from graft-versus-host reaction
  • FIG. 7 shows the detection of LOX and NRAGE in the skin of a patient presenting with a cancer of basocellular or spinocellular type
  • FIG. 8 shows the detection of LOX and NRAGE in the skin of a patient suffering from lichen planus
  • FIG. 9 shows the identification of the location of LOX and NRAGE in the skin of a patient suffering from psoriasis
  • FIG. 10 shows the identification of the location of LOX and NRAGE in the skin of a patient suffering from eczema.
  • FIG. 11 shows the global labelling of the reconstructed skin with Evans blue (red epidermal layers, blue cells, red fibers of the dermal substrate, dermo-epidermal junction in dotted lines).
  • FIG. 12 shows the immuno-histochemical detection of cyto-keratin 10 on the reconstructed skin (cells strongly expressing the red label, the nuclei in blue, the dermo-epidermal junction in dotted lines).
  • FIG. 13 shows the immuno-histochemical detection of transglutaminase on the reconstructed skin (cells strongly expressing the red label, the nuclei in blue, the dermo-epidermal junction in dotted lines).
  • the invention initially consisted in searching for potential partners for LOX in keratinocytes.
  • the yeast two-hybrid technique (Y2H) was employed.
  • the Y2H system makes it possible to identify and characterize interactions between a “lure” protein and “target” potential partners.
  • the lure was the mature region of LOX fused to the DNA binding domain (BD) of the Ga14 gene.
  • the target or targets were the genes encoded by a complementary DNA library of human keratinocytes, fused to the activation domain (AD) of the Gal4 gene.
  • Interaction of the domains of the lure (LOX) with the domains encoded by a library sequence allows binding and activation of the Gal4 promoter, which controls different genes that confer auxotrophy to AH109 yeasts, enabling growth on deficient medium and activation of the galactosidase activities.
  • a gene coding for a protein that was a candidate partner for LOX was thus identified by virtue of this technique for the possible selection of interactants: the intracellular protein Melanoma Associated Antigen D1 (MAGE-D1) or NRAGE ( FIG. 1 ).
  • the cDNA library of normal human skin keratinocytes in vector pGAD-10 was used.
  • the lure chosen for this screening was LOXmat encoded by the human cDNA region of the LOX enzyme without the signal peptide or the pro-region.
  • the nucleotide sequence was inserted in phase behind the DNA binding domain (BD) of the GAL4 transcription factor in vector pBD-Gal4 Cam.
  • the LOX fragment +494 to +1254 to be inserted was amplified by PCR (polymerase chain reaction), and inserted behind the Gal4 binding domain in vector pGal4-BD, using the following primers:
  • the amplified fragment was initially introduced into vector TOPO and then inserted into pGa14-BD.
  • the AH109 yeasts were transformed by lure plasmid pBD-LOXmat.
  • the transformed yeasts are screened: 6.88 ⁇ 10 6 cfu (growth in deficient medium) in the AH109 yeast, 80 selected clones growing in adenine, histidine, tryptophan and leucine deficient medium, 23 of which were retained for their strong growth.
  • Hela cells are transfected on the one hand by plasmids pAct and pAct-NRAGE and on the other hand by pBind and pBind-LOXmat in the presence of lipofectamine.
  • the luciferase activity is tested after 48 h and the results are expressed as the ratio of luciferase activity to control (empty pBind vector).
  • the experiments are performed in triplicate; the mean of the results obtained is shown in FIG. 2 .
  • Example 1 The hLOX sequence used in Example 1 for yeast two-hybrid screening was inserted in phase behind the Gal4 binding domain in the pBind vector (Promega, Madison, USA) for the mammalian two-hybrid interactions.
  • the NRAGE prey was inserted behind the VP16 Gal4 activation domain in the pAct vector (Promega). It starts at amino acid 152 (nucleotide 458).
  • Cos7 cells are co-transfected for the LOX genes (human complete, human mature region and murine mature region) by the construct pcLOX32-V5His (LOX), pcLOX36-V5His or pcLOX27-V5His and for the NRAGE gene by pNM3-HA (complete NRAGE) or pNM7-HA (IRD region).
  • the transfection is performed in Petri dishes 100 mm in diameter using lipofectamine. After 48 h the transfected cells are lyzed in 500 ⁇ l of lysis buffer.
  • the proteins of the cell lyzates are incubated with either anti-V5 or anti-HA; the anti-V5 or anti-HA monoclonal antibody is added to the cell lyzate at a rate of 1/250 over 1 h 30 min at 4° C., with shaking.
  • the immune complexes (ICs) formed in this way are precipitated with protein G-Sepharose over 1 h at 4° C., with shaking. After three 10-minute washes in lysis buffer and elution in SDS-PAGE buffer, all the ICs are subjected to electrophoresis on a 10% SDS-PAGE gel, followed by Western blotting, Transfer of the ICs to a PVDF (polyvinylidene fluoride) membrane is followed by development:
  • PVDF polyvinylidene fluoride
  • the final detection is effected by chemoluminescence of the HRP.
  • NRAGE NM3-HA
  • LOX 32H human complete form of LOX
  • LOX 36H mature human LOX
  • LOX 27H mature murine LOX
  • the identification was effected in a model of reconstructed skin (MIMESKIN®, Engelhard Lyon, France) prepared from a dermal substrate (collagen/glycosaminoglycans/chitosan, MIMEDISC®, Engelhard Lyon, France) inoculated with normal human fibroblasts, on whose surface normal human keratinocytes were deposited.
  • the samples After 45 days of culture, allowing differentiation of the keratinocytes by exposure at the air-liquid interface, the samples are fixed in Bouin's fixative or formaldehyde and then included in paraffin.
  • anti-NRAGE antibody (goat polyclonal IgG),
  • the immune complexes are detected with a peroxidase-conjugated anti-rabbit IgG using diaminobenzidine as substrate, followed by counterstaining with hematoxylin.
  • FIG. 3 shows the immunohistochemical detection of LOX and NRAGE on the reconstructed skin.
  • the position of the dermo-epidermal junction is indicated by a continuous line, the position of the dermal substrate is indicated by an arrow and the location of the keratinocytes is indicated by an arrowhead.
  • LOX appears from the basal cell layer upwards, persists in the suprabasal cell layers and progressively disappears in the differentiated layers.
  • the expression of NRAGE is slightly shifted; it asserts itself from the non-proliferative suprabasal cell layers upwards and intensifies greatly in the granular and horny layers, where LOX is no longer expressed.
  • the inventors thus demonstrated that the LOX and NRAGE proteins are expressed in the epidermis, with a co-location zone in the prickle cell layers.
  • Frozen sections of samples of normal human skin originating from surgical resection are prepared.
  • the anti-LOX and anti-NRAGE primary antibodies of Example 4 were used to detect the expression of LOX and NRAGE.
  • the secondary antibodies used are:
  • a negative control was prepared in the absence of primary antibodies.
  • the dual labeling was observed using a ZEISS AXIOPLAN 2 LSM510 confocal upright microscope and the images were acquired using ZEISS LSM5 Image Browser software.
  • FIG. 4 shows the immunodetection of LOX and NRAGE in normal human skin by confocal microscopy.
  • Example 4 The observations confirm the results of Example 4, further illustrating the co-location of LOX and NRAGE in the prickle cell layers of the epidermis of no mal human skin, with an almost perfect juxtaposition of the expression of LOX and NRAGE.
  • the observations at the cellular level show that LOX and NRAGE appear at the periphery of the cell (in the submembranous peripheral zone), NRAGE also appearing in the cytoplasm.
  • the inventors thus demonstrated fulfilment of the conditions for the direct interaction they had identified in vitro to be able to take place in the epidermis in the zone where both LOX and NRAGE are located.
  • Example 4 An immunohistological study using the protocol described in Example 4 was carried out on sections of human skin originating from 2 donors from different age groups (under 20 years and over 60 years).
  • FIG. 5 shows the labeling performed on the skin of a 91-year-old donor. The observations show a hypoproliferative epidermis that is very thin (reduced to a few cell layers) and hyperkeratinized.
  • NRAGE is strongly expressed, is located in the cytoplasm and appears from the first suprabasal cell layer upwards, without an expression gradient.
  • Example 4 An immunohistological study using the protocol described in Example 4 was carried out on sections of human skin from patients suffering from graft-versus-host reaction (GVH). This study involved 5 different donors.
  • GVH graft-versus-host reaction
  • FIG. 6 identifies an almost total disappearance of LOX in the epidermis (without modification of its expression in the dermis) and a very pronounced presence of NRAGE, which is located in the cytoplasm and appears from the first suprabasal cell layers upwards.
  • Example 4 An immunohistological study using the protocol described in Example 4 was carried out on samples of skin from patients presenting with a basocellular or spinocellular cancer.
  • FIG. 7 shows the following in both types of cancer studied:
  • Example 4 An immunohistological study using the protocol described in Example 4 was carried out on sections of skin from 3 patients suffering from lichen planus.
  • FIG. 8 shows a very large decrease in the expression of LOX in the epidermis, or even its total absence. This disturbance correlates with an irregularity in the expression of NRAGE in the epidermis.
  • Example 4 An immunohistological study using the protocol described in Example 4 was carried out on sections of skin from 5 patients suffering from psoriasis.
  • FIG. 9 is representative of all the sections observed and shows a very strong expression of LOX and a moderate presence of NRAGE in the epidermis, with a more or less homogeneous labeling of the zones in question, showing no expression gradient.
  • LOX is expressed essentially in the lower part of the epidermis and NRAGE solely in its upper part, the expression of the proteins thus being shifted without the overlap zone normally observed in healthy skin.
  • NRAGE is observed only in the cytoplasm and not in the submembranous peripheral zone.
  • Example 4 An immunohistological study using the protocol described in Example 4 was carried out on sections of skin from patients suffering from eczema.
  • FIG. 10 shows a very strong pericellular expression of LOX in the epidermis.
  • NRAGE is weakly expressed and is observed only in the cytoplasm, which reflects a loss of co-location at the cellular level.
  • TUNEL terminal deoxynucleotidyl transferase mediated dUTP nick end labeling
  • the first step consists in labeling the DNA breaks with TdT (terminal deoxynucleotidyl transferase), which catalyzes the polymerization of the fluorescein-labeled nucleotides at the free 3′-OH end of the DNA.
  • the second step consists in detecting the incorporated fluorescein with an alkaline phosphatase-conjugated anti-fluorescein antibody after incubation with the substrate for the latter.
  • the experiment is validated by means of a positive control obtained by fragmentation of the DNA with a DNase solution, and a negative control obtained by depositing phosphate buffer.
  • thermal shock induces pronounced cellular apoptosis
  • inhibition of the enzymatic activity of LOX by ⁇ -APN causes a substantial increase in apoptosis.
  • the keratinocytes are amplified e.g. in K-SFM (keratinocyte serum-free medium), supplemented with antibiotics, up to the third passage at 37° C. under 5% CO 2 .
  • K-SFM Keratinocyte serum-free medium
  • the cells are inoculated into 96-well plates, e.g. at a rate of 40,000 cells per cm 2 , and cultivated to about 80% confluence.
  • the cells are then cultivated in hypercalcium medium (1.7 mM CaCl 2 at 37° C. under 5% CO 2 ) to induce differentiation of the cells.
  • the starting material used to prepare the active principles in the case of plants (preferably roots, stems, barks, flowers, fruits, seeds, germs, gums, exudates, leaves or whole plants) or proteins, may or may not be sterilized by radiation, for example beta or gamma radiation preferably at a dose of 5 kGy, and is then reduced to powder if necessary, for example by grinding at room temperature.
  • the powder is then dispersed at a rate of 2 to 5% (weight/weight) of powder, preferably 5%, either in a polar solvent, e.g.
  • water or butylene glycol, and/or a mixture of polar solvents advantageously a mixture of water and an alcohol, glycol or polyol (such as ethanol, glycerol, butylene glycol and other glycols, xylitol, etc.) in variable proportions, and preferably a 75/25 or 50/50 water/butylene glycol mixture, or in an apolar solvent, e.g. an alkane, or a mixture of apolar solvents, or in a mixture of polar and apolar solvents.
  • an apolar solvent e.g. an alkane
  • a mixture of apolar solvents e.g. an alkane
  • the sample is clarified by decantation or centrifugation and then filtered, preferably on a 0.45 ⁇ m or 0.22 ⁇ m filter.
  • the starting material used to prepare the active principles in the case of characterized molecules (e.g. molecules obtained by synthesis or hemisynthesis, biological molecules obtained by purification), is diluted in a solvent, preferably water or dimethyl sulfoxide (in a concentration preferably of between 10 ⁇ 6 M and 10 ⁇ 2 M and particularly preferably in the order of 10 ⁇ 4 M, or preferably of between 1% weight/weight and 5% weight/weight, depending on the molecules).
  • a solvent preferably water or dimethyl sulfoxide (in a concentration preferably of between 10 ⁇ 6 M and 10 ⁇ 2 M and particularly preferably in the order of 10 ⁇ 4 M, or preferably of between 1% weight/weight and 5% weight/weight, depending on the molecules).
  • a solvent preferably water or dimethyl sulfoxide
  • the active principles obtained by one of the methods described above are then tested in a final concentration preferably of between 0.01% volume/volume (v/v) and 10% (v/v) and advantageously of between 0.1% and 1% (v/v), for example at 1% (v/v).
  • Incubation in the presence of the cells is advantageously carried out for 24 hours in hypercalcium K-SFM without growth factors.
  • the cells are frozen dry at ⁇ 80° C. after being rinsed in phosphate buffer pH 7.4.
  • the total RNAs are extracted with the SV Total RNA Isolation System (Promega, Meylan, France) for the conditions of 96-well plates according to the manufacturer's protocol.
  • the gene expression is modified by real-time RT-PCR, measuring the expression of each gene relative to actin (housekeeping gene) and expressed in % of the untreated negative control.
  • Quantitative real-time RT-PCR Quantitative real-time RT-PCR
  • RNAs at 5 ng/ ⁇ l 10 ⁇ l of total RNAs at 5 ng/ ⁇ l are added to 40 ⁇ l of PCR mix (composed of 25 ⁇ l of SYBR Green Buffer Mix 2 ⁇ , 0.5 ⁇ l of enzyme mix, a final concentration of 0.5 ⁇ M of sense primer and a final concentration of 0.5 ⁇ M of antisense primer, and RNase and DNase free water qsp 40 ⁇ l).
  • the RT-PCR proceeds in different steps comprising retrotranscription at 50° C., 30 min, polymerase activation at 95° C., 15 min, and execution of the PCR cycles (95° C., 15 s; 60° C., 30 s; 72° C., 30 s) ⁇ 50 cycles.
  • the percentage stimulation or inhibition is expressed relative to the untreated control (in the absence of test substance).
  • C(T)gene ⁇ x>> denotes the number of cycles necessary to reach the fluorescence threshold of 0.01-0.05 of the gene ⁇ x>>.
  • the ephedra extract is obtained by extraction of the whole plant, especially with a polar solvent such as water or a water/butylene glycol mixture (e.g. 75/25 or 50/50), preferably water.
  • a polar solvent such as water or a water/butylene glycol mixture (e.g. 75/25 or 50/50), preferably water.
  • the hop extract is obtained by extraction of the cones, especially with a polar solvent such as water or a water/butylene glycol mixture (75/25 or 50/50), preferably water.
  • a polar solvent such as water or a water/butylene glycol mixture (75/25 or 50/50), preferably water.
  • the soya extract is obtained by extraction of the seed, especially with a polar solvent such as water or a water/butylene glycol mixture (75/25 or 50/50), preferably water.
  • a polar solvent such as water or a water/butylene glycol mixture (75/25 or 50/50), preferably water.
  • the ephedra extract can be used e.g. in the treatment of diseases such as cancers, preferably cutaneous epithelial cancers (basocellular or spinocellular), or lichen planus, or optionally in the treatment of certain cutaneous manifestations of GVH, or else to reduce the effects of ageing on the skin.
  • diseases such as cancers, preferably cutaneous epithelial cancers (basocellular or spinocellular), or lichen planus, or optionally in the treatment of certain cutaneous manifestations of GVH, or else to reduce the effects of ageing on the skin.
  • the soya extract can be used e.g. in the treatment of diseases such as cancers, like cutaneous epithelial cancers (basocellular or spinocellular), GVH or lichen planus, or for combating or preventing ageing.
  • diseases such as cancers, like cutaneous epithelial cancers (basocellular or spinocellular), GVH or lichen planus, or for combating or preventing ageing.
  • the soya extract can be used especially for combating cell hyperproliferation.
  • the hop extract can be used e.g. in the treatment of diseases such as cancers, like cutaneous epithelial cancers (basocellular or spinocellular), eczema or psoriasis.
  • diseases such as cancers, like cutaneous epithelial cancers (basocellular or spinocellular), eczema or psoriasis.
  • the hop extract can be used especially for combating cell hypo-proliferation.
  • the soya extract and hop extract can be used in association e.g. in the treatment of diseases such as cancers, like cutaneous epithelial cancers (basocellular or spinocellular), or lichen planus.
  • diseases such as cancers, like cutaneous epithelial cancers (basocellular or spinocellular), or lichen planus.
  • the soya extract and ephedra extract can be used in association e.g. in the treatment of diseases such as cancers, like cutaneous epithelial cancers (basocellular or spinocellular), or lichen planus.
  • diseases such as cancers, like cutaneous epithelial cancers (basocellular or spinocellular), or lichen planus.
  • the hop extract and ephedra extract can be used in association e.g. in the treatment of diseases such as cancers, like cutaneous epithelial cancers (basocellular or spinocellular), or lichen planus.
  • diseases such as cancers, like cutaneous epithelial cancers (basocellular or spinocellular), or lichen planus.
  • the experimental conditions used to obtain cells at 80% confluence are identical to those described in Example 13. Differentiation takes place in the presence of calcium (1.7 mM CaCl 2 ) and active principle. An analysis is performed by Q-RT-PCR (method described in Example 13) after 2, 3 and 4 days of incubation.
  • the cinnamon extract is obtained by extraction of the bark, especially with a polar solvent such as water or a water/butylene glycol mixture (75/25 or 50/50), preferably water.
  • a polar solvent such as water or a water/butylene glycol mixture (75/25 or 50/50), preferably water.
  • the cinnamon extract can be used e.g. in the treatment of diseases such as psoriasis.
  • the cinnamon extract and soya extract (whose effect was detected in the previous Example) can be used in association e.g. in the treatment of diseases such as certain cutaneous manifestations of GVH, eczema or psoriasis, or else for reducing the effects of ageing on the skin.
  • the actives were tested on normal human keratinocytes from young subjects, obtained by enzymatic extraction of human biopsies collected after surgical resection and cultivated as monolayers in a K-SFM (keratinocyte serum free medium with supplements) defined medium, supplemented with antibiotics at 37° C. under 5% CO 2 .
  • K-SFM keratinocyte serum free medium with supplements
  • the cells are sown at the second passage in 24-well plates, for example with an amount of 30 000 cells per cm 2 and grown until about 95% confluence.
  • the cell carpets are rinsed with phosphate buffer at pH 7.4 preferably with calcium and magnesium, before being put into contact with the active ingredients to be tested or the reference positive controls diluted in the K-SFM medium, prepared without supplements but with antibiotics.
  • Active ingredients of different origins are tested from 0.1% volume/volume (v/v) to 1% (v/v).
  • An active ingredient from vegetable origin is tested for example at 1% (v/v) and a synthesized molecule is tested for example at 0.1% (v/v)
  • the active ingredient from vegetable origin are extracts which are obtained by macerating plants (preferably roots, rhizomes, stems, barks, flowers, fruit, seeds, germs or leaves) at 2-5% (w/w) in a solvent or a mixture of solvents, advantageously a 100:0 to 0:100 water/(alcohol, glycol or polyol) (such as ethanol, glycerol, butylene glycol and other glycols, xylitol, etc . . . ) mixture.
  • the obtained extracts are then filtered or distilled in order to recover the soluble fraction which is then filtered at 0.45 ⁇ m preferably.
  • the biotechnological hydrolysates are obtained by fermentation of vegetable extracts in the presence of microorganisms, advantageously from the Lactobacillus or Saccharomyces family. These hydrolysates are then preferably filtered to 0.45 ⁇ m.
  • the incubation is advantageously performed for 24 hours at 37° C. under 5% CO 2 , in a K-SFM medium without growth factors but with antibiotics.
  • the negative controls are either the culture medium by itself, or the culture medium containing from 0.1% (v/v) to 1% (v/v) of the solvent used during the method for extracting the tested extracts.
  • the reference positive control used for inducing cell differentiation is a solution of calcium chloride (CaCl 2 - final concentration of 1.7 mM).
  • the untreated cells (NT control) are dry frozen at ⁇ 80° C. after rinsing with pH 7.4 phosphate buffer. After treatment for 24 hrs in the presence of actives or controls, the cells are dry frozen at ⁇ 80° C. after rinsing with pH 7.4 phosphate buffer.
  • the total RNAs are extracted by means of the SV total RNA isolation system (Promega, Meylan, France) for the conditions of 24-well plates according to the protocol of the manufacturer.
  • Modification of the expression of the genes is achieved by real time RT-PCR which measures the expression of each gene relative to actine (housekeeping gene) and expressed as a % of the untreated negative control (NT).
  • Quantitative Real Time RT-PCR Quantitative Real Time RT-PCR
  • RNAse-free and DNase-free water 10 ⁇ l of 5 ng/ ⁇ l total RNAs are added to 40 ⁇ l of PCR mix (consisting of 25 ⁇ l of SYBR Green Buffer Mix 2 ⁇ , 0.5 ⁇ l of enzyme mix, final 0.5 ⁇ M of sense primer and final 0.5 ⁇ M of antisense primer, RNAse-free and DNase-free water qsp 40 ⁇ l).
  • RT-PCR is performed in different steps including retrotranscription at 50° C., 30 min, activation of polymerase at 95° C., 15 min, performing PCR cycles, (95° C.-15 s, 60° C.-30s, 72° C.-63s, 78° C.-30s) ⁇ 50 cycles.
  • C(T)gene “LOX” meaning the number of cycles required for reaching the fluorescence threshold of 0.01-0.05 of the “LOX” gene.
  • cDNA amounts of each test are relative to the amount of cDNA of actine and then to the negative controls (NT).
  • the results are considered as significative when the measured effect is a modulation by a factor of about 2 (stimulation) or 0.5 (inhibition).
  • NT negative controls
  • the results are considered as significative when the measured effect is a modulation by a factor of about 2 (stimulation) or 0.5 (inhibition).
  • 30 correspond to these criteria under the defined conditions.
  • the reconstructed skin model is in particular achieved according to the following protocol:
  • the active ingredient is advantageously diluted in the culture media described above at 0.5 and 1% and used 3 days after the respective sowing of fibroblasts and keratinocytes (i.e. days 3 to 21 and days 24 to 42).
  • the positive control is preferably made by adding calcium chloride with a final concentration of 1.5 mM during the emerged phase of the reconstructed skins (i.e. day 28-42) in order to stimulate epidermal differentiation.
  • the samples are frozen, included into a thermosensitive resin, and then cryo-cut at 5 ⁇ m.
  • the active ingredient induces the same type of epidermal differentiation as the positive control with a dose-effect relationship.
  • the treated skins have a larger number of keratinocyte layers expressing transglutaminase, in particular in the so-called granular layer.
  • the obtained labelling is more intense and more defined as illustrated in the magnification of the epidermal portion. Quantification clearly demonstrates induction of the protein form after treatment, in particular at 1% active ingredient concentration (2-fold induction).
  • FIG. 11 shows the global labelling of the reconstructed skin with Evans blue (red epidermal layers, blue cells, red fibers of the dermal substrate, dermo-epidermal junction in dotted lines).
  • the active ingredient induces the same type of epidermal differentiation as the positive control with a dose-effect relationship.
  • the treated skins have a larger number of differentiated keratinocyte layers and in particular, an increase in the thickness of the so-called granular layer is noted.
  • an interesting effect was also obtained on the fibroblast density at the dermas treated with ephedra extract.
  • FIG. 12 shows the immuno-histochemical detection of cyto-keratin 10 on the reconstructed skin (cells strongly expressing the red label, the nuclei in blue, the dermo-epidermal junction in dotted lines).
  • the active ingredient induces the same type of epidermal differentiation as the positive control with a dose-effect relationship.
  • the treated skins have a larger number of keratinocyte layers expressing cytokeratin 10 and in particular in the so-called granular layer.
  • FIG. 13 shows the immuno-histochemical detection of transglutaminase on the reconstructed skin (cells strongly expressing the red label, the nuclei in blue, the dermo-epidermal junction in dotted lines).
  • the toxicology tests were performed on the compound obtained according to Example 2, incorporated at a concentration of 10% in a 0.5% xanthan gel, by carrying out an ocular evaluation in the rabbit, by studying the absence of abnormal toxicity by means of a single oral administration in the rat, and by studying the sensitizing power in the guinea-pig.
  • the preparations described above are applied undiluted in a dose of 0.5 ml to the skin of 3 rabbits according to the method recommended by the OECD directive relating to the study of “the acute irritant/corrosive effect on the skin”.
  • the preparations described were given in a single oral administration in a dose of 5 g/kg body weight to 5 male rats and 5 female rats according to a protocol based on OECD directive no. 401 of 24 Feb. 1987 and adapted to cosmetic products.
  • the LD 0 and LD 50 are found to be above 5000 mg/kg. The preparations tested are not therefore classified as dangerous preparations on ingestion.
  • the preparations are classified as non-sensitizing on contact with the skin.

Abstract

The invention relates to the use of a substance for restoring normal co-expression and interaction between the LOX and NRAGE proteins. The invention relates in particular to the use of an effective amount of at least one substance that modulates the expression and/or activity of the LOX of SEQ ID NO: 1 and/or that modulates the expression and/or activity of the NRAGE of SEQ ID NO: 2 for modulating the interaction between the LOX and NRAGE proteins to regulate the balance between the cellular proliferation, differentiation and apoptosis, particularly in cases where the balance between these phenomena is disturbed, and especially in cases where the interaction between LOX and NRAGE is absent or altered. The invention makes it possible to treat skin ageing, lichen planus, graft-versus-host reaction (GVH), eczema, psoriasis and a cancer, particularly an epithelial cancer and more particularly a cutaneous epithelial cancer of basocellular or spinocellular type.

Description

    STATE OF THE ART 1) Background
  • In multicellular organisms, homeostasis is maintained by a balance between cell proliferation, cell differentiation towards a defined function, and programmed or unprogrammed cell death.
  • The skin, for example, can be considered as an organ of which one specific function is to protect the organism from external aggressions. Impermeability to the outside is assured by a highly keratinized corneal layer composed of dead cells. The rhythm of the life of epidermal cells, or keratinocytes, is therefore governed by a succession of changes leading to this differentiation into keratinized and dead cells: (1) maintenance of initiator cells (“stem” cells of the basal cell layer of the epidermis), (2) asymmetric division (each stem cell giving two sister cells), (3) differentiation (formation of the suprabasal cell layers of the epidermis from one of the “initiator sister” cells), (4) acquisition of a resistance to apoptotic cell death, (5) change into cornified cells, and (6) programmed cell death of the upper cell layers of the epidermis. Any modification of this balance can be pathological.
  • Those skilled in the art are familiar with several biological mechanisms and molecules which take account of certain aspects of the cellular phenomena of proliferation, differentiation and apoptosis. However, no data give any real information about any one mechanism which would link these three phenomena and which would thus constitute a target on which to act with a view to controlling and regulating them.
  • 2) LOX (Protein)
  • LOX belongs to the family of the lysyl oxidases (LOs), which are amine oxidases dependent on copper. 5 LO genes have been characterized to date: LOX LOXL, LOXL2, LOXL3 and LOXL4. Where LOX is known for its role in the crosslinking of collagen ex vivo and in vivo, LOXL is clearly associated with the homeostasis of elastic fibers, The in vivo role of the other isoforms is not known.
  • LOs are synthesized by a variety of cells such as fibroblasts, smooth muscle cells, endothelial cells and keratinocytes. This enzyme is therefore present in numerous tissues such as the skin, the liver, the kidneys, the spleen and the aorta, at both extracellular and intracellular levels.
  • 2.1) Stabilization of the Extracellular Matrix—Extracellular Role
  • The extracellular role of LOX, which is now well known, consists in stabilizing the extracellular matrix (ECM) of connective tissues. The actual function of LOX is to crosslink fibrillar collagens and elastin. To do this, it catalyzes the oxidative deamination of lysyl and hydroxylysyl residues of fibrillar procollagen molecules and tropoelastin, this reaction being accompanied by the release of ammonia and hydrogen peroxide. The aldehyde residues formed then condense spontaneously with adjacent aldehyde or amine groups to form intramolecular and intermolecular bonds. These condensation reactions give rise to the bridging found in collagen and elastin fibers. LOX and LOs are therefore studied in biomedical fields that involve a change in the ECM (ageing, fibroses, cancer, healing, osteoarticular and cardiovascular diseases, angiogenesis).
  • It has been demonstrated in vitro that organic nitriles are irreversible LOX inhibitors. Thus β-aminopropionitrile (β-APN) binds to the active site of LOX in competition with alkylamine substrates; LOs use β-APN as substrate, thereby forming a Schiff base but without releasing the aldehyde product, which blocks the active site covalently without having an effect on synthesis. On this basis the inventors have described the use of different lysyl oxidase inhibitors, including β-APN, for avoiding the dedifferentiation of certain cell types (chondrocytes, etc.) which occurs systematically when these cells are cultivated (Farjanel et al., French patent 01.10443, CNRS, Use of lysyl oxidase inhibitors for cell culture and tissue engineering, filed on 3 Aug. 2001, publication no.: 2 828 206). However, said document relates to lysyl oxidases as a whole and not specifically to the LOX isoform; furthermore, it relates exclusively to inhibition of the dedifferentiation of cells cultivated in vitro, providing no teaching on the role of lysyl oxidases, or a fortiori of LOX, in maintenance of the balance between proliferation, differentiation and apoptosis, nor does it provide any teaching on novel partners or substrates for lysyl oxidases or LOX.
  • 2.2) Intracellular Role
  • The intracellular role of LOX, and of LOs in general, is less well known. Thus, although it is acknowledged that LOX participates in the regulation of cell development, differentiation, mobility or senescence (Csiszar, Lysyl oxidases: A novel multifunctional amine oxidase family, Nucleic Acid Research and Molecular Biology, 2001, 70, 2-28), the underlying molecular mechanisms are not elucidated.
  • 2.2.1) LOX and Cellular Homeostasis
  • The possible involvement of LOX in the regulation of cellular homeostasis (maintenance of a physiological balance between proliferation, differentiation and apoptosis) is commonly accepted (Jeay et al., Lysyl oxidase inhibits Ras-mediated transformation by preventing activation of NF-KB, Mol. Cell. Biol., 2003, 23, 2251-2263), but the underlying mechanisms are still unknown to those skilled in the art.
  • At the present time, those skilled in the art are thus in possession of a few data indicating a possible relationship between LOX and differentiation, on the one hand, and LOX and cell proliferation/transformation, on the other. However, the prior art does not refer to a possible relationship between LOX and apoptosis.
  • 2.2.2) LOX and Epidermal Differentiation
  • LOX has been located in the epidermis, its expression being regulated as a function of the level of differentiation of keratinocytes (Noblesse et al., Lysyl oxidase-like and lysyl oxidase are present in the dermis and epidermis of a skin equivalent and in human skin and are associated to elastic fibers, J. Invest. Dermatol., 2004, 122, 621-630). However, the studies carried out hitherto have not made it possible to define the role of LOX in keratinocytes or to search for its partners in these cells, which make little or no collagen or elastin.
  • 2.2.3) LOX and Cell Transformation
  • The LOX gene is clearly associated with maintenance of the non-tumoral phenotype of cells.
  • Two hypotheses have been put forward to explain this role of LOX. The first hypothesis suggests that crosslinking of the ECM induces a three-dimensional environment that favors maintenance of the non-tumoral state. This hypothesis is supported by the fact that LOX and LOXL are no longer expressed when cancers become invasive, whereas they are present in cancers in situ (Peyrol et al., Lysyl oxidase gene expression in the stromal reaction to in situ and invasive ductal breast carcinoma, Am. J. Pathol. Feb., 150(2), 497-507, 1997). The other hypothesis concerns the intracellular roles of LOs on substrates which those skilled in the art are yet to discover (Li et al., Localization and activity of lysyl oxidase within nuclei of fibrogenic cells, Proc. Natl. Acad. Sci. USA, 1997, Nov. 25, 94(24), 12817-22).
  • It has in fact been shown that the LOX enzyme is a suppresser of the ras oncogene and that somatic mutations in the gene are associated with various cancers (Contente et al., Expression of gene rrg is associated with reversion of NIH 3T3 transformed by LTR-c-H-ras, Science, 1990, 249, 796-798; Csiszar et al., Somatic mutations of the lysyl oxidase gene on chromosome 5q23.1 in colorectal tumors, Int. J. Cancer, 2002, 97, 636-642). Recent studies show that the low level of expression of LOX in fibroblasts transformed by ras is due to the activity of the FGF-2 autocrine growth factor and that the antitumoral drug suramine makes it possible to reinduce the expression of the enzyme (Palamakumbura et al., Autocrine growth factor regulation of lysyl oxidase expression in transformed fibroblasts, J. Biol. Chem., 2003, Aug. 15, 278(33), 30781-7; Epub 2003 Jun. 4). It has been shown elsewhere that the re-expression of LOX in these fibroblasts inhibits their growth in soft agar by acting on the NF-kB signaling pathway via regulation of the location of the AKT protein (Jeay et al., Lysyl oxidase inhibits Ras-mediated transformation by preventing activation of NF-B, Mol. Cell. Biol., 23, 2251-2263, 2003).
  • As in the case of other tumor suppressors, LOX probably acts according to the availability and regulation of its cellular substrates and partners, but the latter are still unknown to those skilled in the art.
  • 2.2.4) Apoptosis
  • The term apoptosis is used to describe a particular form of cell death whose morphological characteristics differ from those of necrosis.
  • Apoptosis is a process of programmed cell death that requires the acquisition of caspases. In the course of this process the cells acquire particularly remarkable morphological characteristics, such as condensation of the chromatin and fragmentation of the nucleus, leading to their self-destruction and their elimination from the tissue without damaging the adjacent cells.
  • Apoptosis corresponds to the natural death of cells in the course of their development or during homeostasis.
  • The prior art provides information about a number of agents and mechanisms that are involved in regulating the cell cycle via the cellular processes of apoptosis and differentiation,
  • 3) NRAGE (Protein)
  • Thus we are particularly familiar with one family of proteins that are involved in regulating the cell cycle, differentiation and apoptosis. It is the family of the MAGE (melanoma associated antigen) proteins, one of whose members, the NRAGE protein, which is also called “neurotrophin-receptor-interacting MAGE homolog”, is particularly known for its proapoptotic role via the neurotrophic factor NGF.
  • The NRAGE protein comprises 778 amino acids. In the central region it carries a first domain characteristic of the MAGE proteins: the MAGE homology domain (MHD-1), and in the N-terminal region it carries a second domain: MHD-2, present only in certain isoforms. These two domains have zones rich in lysyl residues. Between these two domains there is a region called IRD (interspersed repeat domain), which does not exist in any other protein currently known,
  • The NRAGE protein is involved in controlling apoptosis via different pathways.
  • By interacting with p75NTR, which is the “low affinity” receptor of neutrophilic factors (NGF) or TNF, NRAGE can block the progression of the cell cycle and thus is proapoptotic via the caspase pathway.
  • NRAGE is also proapoptotic by interacting with cytoplasmic inhibitors of apoptotic proteins, IAP.
  • NRAGE can also act directly on the activity of nuclear homeo factors, such as the factors Msx and Dix, which take part in the morphogenic regulation of tissues.
  • Although the expression of NRAGE is ubiquitous, the prior art does not refer to its presence in the skin, nor does it provide any data giving information about a possible relationship between LOX and apoptosis or about a link between LOX and NRAGE.
  • 4) Conclusion on the Prior Art
  • The prior art does not provide the identity of novel partners or substrates for LOX, especially those taking part in maintenance of the cellular balance between proliferation, differentiation and apoptosis; moreover, the prior art does not provide the identity of these novel partners or substrates either in epithelial cells (particularly keratinocytes) or in any other cell type.
  • The prior art gives no information about the possible variations in the expression of LOX in the epidermis (especially LOX expressed by keratinocytes) as a function of age or of the existence or non-existence of exposure to UV or other types of aggression, or in the case of diseases affecting the skin (psoriasis, graft-versus-host reaction, cancers, etc.).
  • The prior art does not describe the involvement of LOX in apoptosis.
  • The prior art does not provide information about the possible presence of NRAGE in the skin, whether it be healthy skin, skin altered by age or UV or subjected to other types of aggression, or diseased skin.
  • The prior art does not offer known models for studying NRAGE in cells of epithelial origin or in the epidermis.
  • The prior art provides no information about any kind of interaction between LOX and NRAGE, irrespective of the tissue.
  • The prior art does not provide a model for identifying active principles capable of modulating the expression of LOX and/or NRAGE in keratinocytes.
  • Furthermore, animal experimentation is currently banned in Europe for certain applications and human experimentation is the subject of ethical debate. It is therefore unacceptable for the inventors to implement a screening method using animals or humans.
  • In the three-dimensional model MIMESKIN® (Coletica, France), LOX is expressed in the epidermis, its expression being regulated as a function of the level of differentiation of the keratinocytes (Noblesse et al., 2004). These studies did not include research into the expression of NRAGE or into the existence of a possible relationship between LOX and apoptosis, so it was not obvious that those skilled in the art would be interested in modulating the expression of LOX and/or NRAGE in order to regulate cellular homeostasis, based on the balance between proliferation, differentiation and apoptosis, in cases where it is disturbed (age, stress, disease), which thus constitutes a novel technical problem.
  • The prior art therefore fails to provide active principles capable of modulating the expression of intracellular partners for LOX (such as NRAGE), which may or may not be associated with modulation of the expression of LOX for the purpose of acting on cell regulation. In this context it is also very difficult to obtain objective criteria for judging the impact of these active principles.
  • OBJECTS OF THE INVENTION
  • The main object of the invention is to solve the technical problems referred to above, especially the technical problem relating to the provision of a method of identifying active principles for improving the interaction between the LOX and NRAGE proteins in cases where the interaction between LOX and NRAGE is absent or altered.
  • In particular the aim of the invention is to regulate the cellular balance between proliferation, differentiation and apoptosis in cases where it is disturbed, as in the case of skin altered by age or UV or subjected to other types of aggression, and/or in the case of pathological situations such as psoriasis, eczema, graft-versus-host reaction, lichen planus and/or cancerous diseases.
  • The invention further relates to the use of an active principle that modulates the expression of LOX and/or NRAGE for regulating the cellular balance between proliferation, differentiation and apoptosis, particularly in cases where it is disturbed, as in the case of skin altered by age or UV or subjected to other types of aggression, and/or in the case of pathological situations such as psoriasis, eczema, graft-versus-host reaction, and cancerous scars and/or diseases. In particular the invention relates to active principles for improving the interaction between the LOX and NRAGE proteins in cases where the interaction between LOX and NRAGE is absent or altered.
  • The invention makes it possible to solve the technical problem that consists in providing a method of locating the expression of NRAGE and following this expression, especially on a cellular model of skin. The invention makes it possible to solve the technical problem that consists in providing a method of diagnosing a state associated with poor regulation of the interaction between LOX and NRAGE.
  • DESCRIPTION OF THE INVENTION
  • The meanings of the terms used throughout the description, as understood by the inventors, are given below:
  • “LOX”: the isoform of the human protein lysyl oxidase, LOX, as defined in particular by the amino acid sequence SEQ ID no. 1;
  • “NRAGE”: the human protein NRAGE as defined in particular by the amino acid sequence SEQ ID no. 2;
  • “modulation of the expression of LOX”: modulation of the gene coding for LOX, especially modulation of the expression of the messenger RNA coding for LOX, but also modulation of the synthesis of LOX from this messenger RNA, as well as modulation of the activity of LOX;
  • “modulation of the expression of NRAGE”: modulation of the gene coding for NRAGE, especially modulation of the expression of the messenger RNA coding for NRAGE, but also modulation of the synthesis of NRAGE from this messenger RNA, as well as modulation of the biological effect of NRAGE.
  • These modulations must make it possible to reinduce a state of balance between proliferation, differentiation and apoptosis in situations where this balance is disturbed.
  • Active principles which are considered to be effective on LOX are preferably those affording a difference of about ±50% in the expression of the mRNA of LOX and/or a difference of about ±15% in the expression of LOX and/or the activity of LOX on a model, comprising at least one cell type exhibiting LOX expression and/or activity, in contact with these active principles, compared with the level of LOX expression and/or activity in a control model (generally without being brought into contact with the active principles).
  • Active principles which are considered to be effective on NRAGE are preferably those affording a difference of about ±50% in the expression of the mRNA of NRAGE and/or a difference of about ±15% in the expression of NRAGE and/or the biological effect of NRAGE on a model, comprising at least one cell type exhibiting NRAGE expression and/or activity, in contact with these active principles, compared with the level of NRAGE expression and/or activity in a control model (generally without being brought into contact with the active principles).
  • Thus, according to a first feature, the present invention relates to the use of an effective amount of at least one substance that modulates the expression and/or activity of at least the LOX protein of sequence ID no. 1, and/or that modulates the expression and/or activity of at least the NRAGE protein of sequence ID no. 2, for the manufacture of a composition for modulating the interaction between LOX and NRAGE in order to regulate the cellular balance between proliferation, differentiation and apoptosis, particularly in cases where the balance between these phenomena is disturbed, and especially in cases where the interaction between LOX and NRAGE is absent or altered.
  • The invention relates to the use of an effective amount of at least one substance that modulates the expression and/or activity of LOX of sequence ID no. 1, and/or that modulates the expression and/or activity of NRAGE of sequence ID no. 2, for the manufacture of a composition for improving the interaction between the LOX and NRAGE proteins in cases where the interaction between LOX and NRAGE is absent or altered.
  • The invention further relates to the use of an effective amount of at least one substance that modulates the expression and/or activity of at least the NRAGE protein of sequence ID no. 2, and that optionally modulates the expression and/or activity of the LOX protein of sequence ID no. 1, for the manufacture of a composition for preventing or treating at least one state in which the cellular balance between proliferation, differentiation and apoptosis is absent or altered.
  • Advantageously, the expression of LOX and/or NRAGE is modulated in epithelial cells, particularly in keratinocytes.
  • Advantageously, the purpose of said substance is to modulate the interaction between the LOX protein and the NRAGE protein, said interaction occurring especially in the IRD domain of the NRAGE protein.
  • Advantageously, the interaction between LOX and NRAGE comprises the polymerization of NRAGE, particularly its dimerization, by LOX-induced enzymatic catalysis.
  • Advantageously, the interaction between LOX and NRAGE involves the production of H2O2 resulting from the catalytic activity of LOX, the H2O2 then activating other molecules, for example neutral sphingomyelinase or NF-kB.
  • Advantageously, the interaction between LOX and NRAGE involves a non-enzymatic activity of LOX, particularly an activity exerted by the pro-region (A22-D169) of LOX.
  • Advantageously, the purpose of the substance is to treat and/or prevent a condition selected from the group consisting of exposure of cells to a stress, particularly exposure of cells to heat, or exposure of cells to radiation, particularly solar radiation, or exposure of cells to a toxic agent, for example a chemical or microbiological agent, skin ageing, lichen planus, graft-versus-host reaction (GVH), eczema, psoriasis and a cancer, particularly an epithelial cancer.
  • Advantageously, the purpose of the substance is to reduce cell proliferation in the case of hyperproliferation, particularly in a cancer, more particularly an epithelial cancer and most particularly a cutaneous epithelial cancer, of basocellular or spinocellular type, psoriasis or eczema.
  • Advantageously, the purpose of the substance is to reduce apoptosis in the epidermis in the case of substantial apoptosis, particularly during skin ageing, exposure of the skin to a stress, particularly exposure to heat, or exposure of the skin to radiation, particularly solar radiation, or exposure of the skin to a toxic agent, for example a chemical or microbiological agent, or graft-versus-host reaction (GVH).
  • Advantageously, the purpose of the substance is to increase cell proliferation in the case of cell hypoproliferation in the epidermis, particularly during ageing, exposure of the skin to heat, exposure of the skin to radiation, particularly solar radiation, or graft-versus-host reaction (GVH).
  • Advantageously, the purpose of the substance is to stimulate the expression of LOX, and optionally to inhibit the expression of NRAGE, during skin ageing, exposure of the skin to a stress, particularly exposure to heat, or exposure of the skin to radiation, particularly solar radiation, or exposure of the skin to a toxic agent, for example a chemical or microbiological agent, or graft-versus-host reaction (GVH).
  • Advantageously, the purpose of the substance is to stimulate the expression and/or activity of NRAGE, and optionally to inhibit the expression and/or activity of LOX, in the epidermis in order to prevent or treat psoriasis or eczema.
  • Advantageously, the purpose of the substance is to stimulate the expression of LOX and NRAGE in order to prevent or treat a cancer, particularly an epithelial cancer and more particularly a cutaneous epithelial cancer, of basocellular or spinocellular type, or lichen planus.
  • Advantageously, said composition is a cosmetic, neutraceutical, dermo-pharmaceutical or pharmaceutical composition.
  • Advantageously, the cellular balance between proliferation, differentiation and apoptosis is the balance between proliferation, differentiation and apoptosis of the keratinocytes.
  • Advantageously, the starting material used to prepare the active principles, in the case of plants (preferably roots, stems, barks, flowers, fruits, seeds, germs, gums, exudates, leaves or whole plants) or proteins, may or may not be sterilized by radiation, for example beta or gamma radiation preferably at a dose of 5 kGy, and is then reduced to powder if necessary, for example by grinding at room temperature. The powder is then e.g. dispersed at a rate of 2 to 5% (weight/weight) of powder, preferably 5%, either in a polar solvent, for example water, an alcohol, a glycol such as butylene glycol, or a polyol, and/or a mixture of polar solvents, advantageously a mixture of water with an alcohol, glycol or polyol (such as ethanol, glycerol, butylene glycol and other glycols, xylitol, etc.) in variable proportions, preferably a 75/25 or 50/50 water/butylene glycol mixture, or in an apolar solvent, for example an alkane, or a mixture of apolar solvents, or in a mixture of polar and apolar solvents. Preferably after stirring, e.g. magnetic stirring, for at least 2 hours, and optionally heating of the solvent, the sample is preferably clarified by decantation or centrifugation and then filtered, preferably on a 0.45 μm or 0.22 μm filter.
  • Advantageously, the starting material used to prepare the active principles, in the case of characterized molecules (e.g. molecules obtained by synthesis or hemisynthesis, biological molecules obtained by purification), is diluted in a solvent, preferably water or dimethyl sulfoxide (in a concentration preferably of between 10−6 M and 10−2 M and particularly preferably in the order of 10−4 M, or preferably of between 1% weight/weight and 5% weight/weight, depending on the molecules). The solution obtained is then optionally filtered, preferably on a 0.45 μm or 0.22 μm filter.
  • Advantageously, the active principles obtained by one of the methods described above are used in a final concentration preferably of between 0.01% volume/volume (v/v) and 10% (v/v) and particularly preferably of between 0.1% and 1% (v/v).
  • Advantageously, said substance is selected from the group consisting of a soya extract, an ephedra extract, a hop extract and a cinnamon extract.
  • According to a second feature, the invention relates to a method of identifying at least one active principle described above, the purpose of which is in particular to modulate the interaction between LOX and NRAGE in order to prevent or treat at least one state in which the cellular balance between proliferation, differentiation and apoptosis is absent or altered, characterized in that it comprises:
  • bringing the active principle into contact with at least one type of living cell capable of expressing the LOX protein (SEQ ID no. 1) and/or the NRAGE protein (SEQ ID no. 2), and
  • analyzing the expression of LOX and/or NRAGE, especially to identify an active principle that modulates the expression and/or activity of LOX and/or NRAGE in order to improve the cellular balance between proliferation, differentiation and apoptosis.
  • The invention further relates to a method of identifying at least one active principle for modulating the interaction between LOX and NRAGE in order to improve the interaction between the LOX and NRAGE proteins in cases where the interaction between LOX and NRAGE is absent or altered, characterized in that it comprises:
  • bringing the active principle into contact with at least one type of living cell capable of expressing the LOX protein (SEQ ID no. 1) and/or the NRAGE protein (SEQ ID no. 2), and
  • analyzing the expression of LOX and/or NRAGE, especially to identify an active principle that modulates the expression and/or activity of LOX and/or NRAGE in order to improve the interaction between the LOX and NRAGE proteins in cases where the interaction between LOX and NRAGE is absent or altered.
  • Advantageously, the balance between the proliferation, differentiation and apoptosis of said living cells capable of expressing the LOX and/or NRAGE protein, or the interaction between the LOX and NRAGE proteins, is absent or altered before they are brought into contact with the active principle.
  • Advantageously, the living cells are epithelial cells, particularly keratinocytes.
  • Advantageously, the method comprises analyzing the expression of the messenger RNAs of LOX and/or NRAGE.
  • Advantageously, the method comprises using quantitative RT-PCR with the following primers in particular:
  • for the LOX gene:
  • Sense
    ACGTACGTGCAGAAGATGTCC
    Antisense
    GGCTGGGTAAGAAATCTGATG
  • for the NRAGE gene:
  • Sense
    TGCACAGACATCAGCAGATGG
    Antisense
    TTCACGGATGATATCTCTCAGC
  • Advantageously, the method comprises analyzing the kinetics of expression of the messenger RNAs, for example by quantitative RT-PCR.
  • The invention further relates to a method for the cosmetic care or therapeutic treatment of a subject in whom the balance between the cellular phenomena of proliferation, differentiation and apoptosis in at least one cell type is disturbed, said method comprising improving the interaction between LOX and NRAGE by the application or administration of a substance that modulates the expression of LOX and/or NRAGE.
  • The invention further relates to a method for the cosmetic care or therapeutic treatment of a subject in whom the interaction between LOX and NRAGE is absent or altered, said method comprising the application or administration of a substance that modulates the expression and/or activity of LOX of sequence ID no. 1, and/or that modulates the expression and/or activity of NRAGE of sequence ID no. 2.
  • The invention further relates to a method for the cosmetic care or therapeutic treatment of a subject presenting with a state in which the cellular balance between proliferation, differentiation and apoptosis is absent or altered, said method comprising the application or administration of a substance that modulates the expression and/or activity of NRAGE, and optionally that modulates the expression and/or activity of LOX.
  • According to a third feature, the invention relates to a process for the preparation of a composition, comprising:
  • the use of a previously defined method of identification for identifying an active principle that modulates the expression and/or activity of LOX and/or NRAGE in order to improve the cellular balance between proliferation, differentiation and apoptosis, and
  • the mixing of the active principle with at least one excipient in order to produce a cosmetic, neutraceutical, dermopharmaceutical or pharmaceutical composition for preventing or treating at least one state in which the cellular balance between proliferation, differentiation and apoptosis is absent or altered.
  • The invention further relates to a process for the preparation of a composition, comprising:
  • the use of a previously defined method of identification for identifying an active principle that modulates the expression and/or activity of LOX and/or NRAGE in order to improve the interaction between the LOX and NRAGE proteins, and
  • the mixing of the active principle with at least one excipient in order to produce a cosmetic, neutraceutical, dermopharmaceutical or pharmaceutical composition for improving the interaction between the LOX and NRAGE proteins in cases where the interaction between LOX and NRAGE is absent or altered.
  • According to a fourth feature, the invention relates to a method of locating NRAGE, comprising the use of at least one anti-NRAGE antibody for detecting and locating the presence of NRAGE, particularly in a model of reconstructed skin comprising at least keratinocytes, or a section of skin, preferably a section of skin originating from a person whose epidermis exhibits a state in which the cellular balance between proliferation, differentiation and apoptosis is absent or altered.
  • According to a fifth feature, the invention relates to the use of an anti-NRAGE antibody for the preparation of a composition for detecting a modulation of the expression of NRAGE at the cellular level, particularly in epithelial cells and preferably in keratinocytes.
  • Advantageously, the purpose of the composition is to detect a state in which the cellular balance between proliferation, differentiation and apoptosis, or the interaction between LOX and NRAGE, particularly in the epidermis, is absent or altered, said state being selected from the group consisting of exposure of cells to a stress, particularly exposure of cells to heat, or exposure of cells to radiation, particularly solar radiation, or exposure of cells to a toxic agent, for example a chemical or microbiological agent, skin ageing, lichen planus, graft-versus-host reaction (GVH), eczema, psoriasis and a cancer, particularly an epithelial cancer and more particularly a cutaneous epithelial cancer, of basocellular or spinocellular type.
  • Advantageously, the composition also comprises an anti-LOX antibody for detecting a modulation of the expression of LOX at the cellular level, particularly in epithelial cells and preferably in keratinocytes.
  • Advantageously, the composition also comprises an anti-LOX antibody for detecting a modulation of the expression of LOX in keratinocytes.
  • Advantageously, the keratinocytes are human keratinocytes.
  • DETAILED DESCRIPTION OF THE INVENTION:
  • Unexpectedly, the inventors have discovered an interaction between LOX and the NRAGE protein. This discovery is fundamental and is the starting point of the present invention. It actually shows LOX to be the protein which, being present at the intersection of the proliferation, differentiation and apoptosis pathways, is capable of controlling cellular homeostasis.
  • The inventors have also discovered that the NRAGE protein is present in particular in the epidermis and is expressed in particular by the keratinocytes. This discovery also enables the interaction between LOX and NRAGE to be made an action target for cosmetic care or treatments for restoring cellular homeostasis in situations where it is disturbed.
  • 1) Discovery and Characterization of the LOX/NRAGE Interaction In Vitro
  • Having previously discovered the presence of LOX in the epidermis (Noblesse et al., Lysyl oxidase-like and lysyl oxidase are present in the dermis and epidermis of a skin equivalent and in human skin and are associated to elastic fibers, J. Invest. Dermatol., 122, 621-630, 2004), the inventors endeavored to determine the role(s) that LOX might play at this level, in the knowledge that its main currently acknowledged role—crosslinking of collagen and elastin—was rather unlikely in these cells, which make little or no collagen or elastin at the level where LOX was located.
  • With this objective in mind, they searched for protein partners for LOX by the yeast two-hybrid technique and a library of normal human skin keratinocyte cDNA was screened using the lure GAL4 BD-hLOXmat in particular, thereby making it possible to identify NRAGE as a potential partner for LOX (cf. Example 1).
  • Using the two-hybrid technique in Hela cells, the inventors then verified that this potential partnership was also observed in mammalian cells (cf. Example 2).
  • The next step consisted in determining whether the identification of this potential partnership between LOX and NRAGE could correspond to a direct physical interaction between these two proteins. This was carried out in particular by a technique involving co-immunoprecipitation of the LOX and NRAGE proteins produced after transfection of their genes into Cos7 cells. The results obtained on the one hand confirmed the physical interaction between LOX and NRAGE and on the other hand made it possible to specify that LOX interacted with a particular region of NRAGE called IRD (interspersed repeat domain), which is a region specific to NRAGE in the family of the MAGE proteins (cf. Example 3).
  • In this context it should be pointed out at this stage that the discovery of the interaction between LOX and NRAGE is of very particular interest when one considers, in combination, on the one hand that the NRAGE protein contains two domains rich in lysyl residues, which could be favorable sites for its dimerization under the catalytic action of LOX, and on the other hand that it is precisely in the form of a dimer that the functionality of NRAGE seems to manifest itself.
  • With this discovery of the existence of a direct physical interaction between LOX and NRAGE in vitro, located in the IRD region of NRAGE, the inventors asked themselves the question whether this interaction also took place in vivo. It is essential to recall at this stage that, in contrast to LOX, the presence of NRAGE in the skin had never previously been described.
  • 2) Identification of the Presence of NRAGE in Normal Human Skin and Reconstructed Normal Human Skin
  • The inventors identified the expression of NRAGE in both the dermis and the epidermis of the skin (cf. Examples 4 and 5).
  • Within the framework of the present invention the inventors used a method of locating the expression of NRAGE in the skin.
  • In the epidermis the inventors demonstrated, unexpectedly, that NRAGE was not expressed homogeneously—as would have been expected of a protein whose presence is generally described as ubiquitous—but in the form of an expression gradient.
  • Reminder of the structure of the epidermis:
  • The epidermis is a stratified epithelium resting on a basal membrane which anchors it to the dermis. Its thickness varies from 60 to 100 μm on average and it consists of 4 continuous layers, from the deepest to the surface, resulting from a progressive differentiation of the keratinocytes:
  • the basal cell layer (single row of cells)
  • the prickle cell layer (5 to 6 monocellular layers)
  • the granular layer (1 to 3 monocellular layers)
  • the horny layer (5 to 10 monocellular layers)
  • Thus NRAGE is absent in the basal cell layer and appears in the first layers of differentiated keratinocytes, the labeling increasing in the horny layers. The labeling of the first suprabasal cell layers of the epidermis corresponds to that observed for LOX, which appears from the basal cell layer upwards. On the other hand, the labeling of LOX decreases in the upper layers of the epidermis, where NRAGE is present, so a distinction is made between the following 3 zones:
  • 1 lower zone of the epidermis, comprising the basal cell layer and the first proliferative suprabasal cell layers, where only LOX is expressed;
  • 1 intermediate zone extending from the first non-proliferative suprabasal cell layers to the first monocellular horny layers, in which LOX and NRAGE are co-expressed;
  • and 1 upper zone of the epidermis, where only NRAGE is expressed.
  • Observations at the cellular level show that LOX and NRAGE appear at the periphery of the cell, particularly in the submembranous region, NRAGE also appearing in the cytoplasm (cf. Examples 4 and 5).
  • The inventors thus identified, for the first time, the presence of NRAGE in the skin, both in the dermis and the epidermis. They also showed that the epidermis comprises a part in which both the LOX and NRAGE proteins are expressed and share the same location at the cellular level, especially in the keratinocytes, said location being the submembranous peripheral zone (NRAGE also being present in the cytoplasm).
  • The inventors thus demonstrated fulfilment of the conditions for the direct interaction they had identified in vitro to be able to take place in the epidermis in the zone where both LOX and NRAGE are located.
  • 3) Identification of Disturbances of the Ex Ression of LOX and/or NRAGE in the Epidermis With Age and in Certain Pathological Situations
  • The inventors also demonstrated, unexpectedly, that skin ageing and a number of pathological situations are accompanied by disturbances of the expression of LOX and/or NRAGE in the epidermis.
  • 3.1) State of LOX and NRAGE in the Epidermis
  • 3.1.1) Skin of Elderly Persons
  • The skin of elderly persons is characterized by a hypoproliferative epidermis that is very thin (reduced to a few cell layers) and hyperkeratinized.
  • The inventors identified the total absence of LOX (detectable by the techniques used) in aged skin. On the other hand, NRAGE is strongly expressed, is located in the cytoplasm and appears from the first suprabasal cell layer upwards, without an expression gradient.
  • Thus the present invention makes it possible to stimulate the expression (and/or activity) of LOX, with or without inhibiting the expression (and/or activity) of NRAGE, so as to reinduce a regulated differentiation zone and/or apoptosis zone, in particular by restoring a LOX and NRAGE co-expression zone. This also thickens the skin, especially the epidermis.
  • Thus the purpose of the present invention is especially to correct or prevent the effects of skin ageing.
  • 3.1.2) Graft-Versus-Host Reaction (GVH)
  • GVH is a disease which can arise following allografts of hemopoietic stem cells. It is associated with the effect of the immune cells (lymphocytes) contained in the graft on the patient's normal organs (especially the skin, liver and digestive tract). In the skin it manifests itself in the form of a maculopapular, pruriginous and inflammatory eruption.
  • Patients suffering from this disease have very thin skin with a highly apoptotic epidermis.
  • The histological study carried out by the inventors identified the absence of LOX and a very pronounced presence of NRAGE, which is located in the cytoplasm and appears from the first suprabasal cell layers upwards.
  • Thus the present invention makes it possible to stimulate the expression (and/or activity) of LOX, with or without inhibiting the expression (and/or activity) of NRAGE, in order to reinduce a LOX and NRAGE co-expression zone, enabling them to interact and resulting in a reduction in the cutaneous manifestations of GVH, this being accompanied especially by a thickening of the skin, particularly the epidermis.
  • Thus the present invention enables patients suffering from GVH to reduce the cutaneous manifestations of this disease.
  • 3.1.3) Lichen Planus
  • Lichen is a skin disease of unknown cause which is characterized by the presence of violet, flat, solid, dry and very pruriginous papules a few millimeters in diameter.
  • The inventors identified a very large decrease in the expression of LOX, or even its total absence, and a very irregular expression of NRAGE. These observations certainly reflect a non-existent or very weak level of interaction between the two proteins studied.
  • Thus the present invention makes it possible to stimulate the expression (and/or activity) of LOX, with or without modulating the expression (and/or activity) of NRAGE, in order to reinduce a LOX and NRAGE co-expression zone, enabling them to interact and making it possible to return to a normal epidermal state.
  • Thus the present invention enables patients suffering from lichen planus to treat, reduce or prevent this disease.
  • 3.1.4) Psoriasis
  • Psoriasis is a chronic skin disease characterized by erythematosquamous lesions. The fundamental feature of the disease is an increase in the rate of multiplication of the keratinocytes, which is responsible for a more rapid renewal and a thickening of the epidermis.
  • Histologically one observes a hyperproliferation of the cells involved in the first steps of terminal differentiation, i.e. an incomplete terminal differentiation, which is associated with the absence or very weak presence of apoptosis.
  • The inventors detected a very strong expression of LOX and a moderate presence of NRAGE with a more or less homogeneous labeling of the zones in question, showing no expression gradient. These characteristics of an intensity of expression that varies from the norm are coupled with much more important anomalies affecting the location of the proteins in question. Thus, in psoriatic skin, LOX is essentially expressed in the lower part of the epidermis and NRAGE solely in its upper part, the expression of the proteins thus being shifted without there being the overlap zone normally observed in healthy skin. At the cellular level NRAGE is observed only in the cytoplasm and not in the submembranous peripheral zone.
  • These observations show that, in psoriasis, although the LOX and NRAGE proteins are both present in the epidermis, they cannot interact directly with one another because they are not physically present in the same place. This absence of interaction results in dysfunctions observed in the epidermis, thus reflecting the role of the interaction between LOX and NRAGE in maintaining epidermal homeostasis.
  • Thus inhibition of the expression (and/or activity) of LOX, and/or optionally stimulation, preferably partial stimulation, of the expression (and/or activity) of NRAGE, makes it possible in particular to obtain an overlap zone of LOX and NRAGE expression in order to reinduce a regulated proliferation zone that makes it possible to reduce hyperproliferation by promoting apoptosis in the keratinocytes.
  • Furthermore, stimulation of the expression (and/or activity) of NRAGE, and/or inhibition, preferably partial inhibition, of the expression (and/or activity) of LOX, makes it possible in particular to obtain an overlap zone of LOX and NRAGE expression in order to reinduce a regulated proliferation zone that makes it possible to reduce hyperproliferation by promoting apoptosis in the keratinocytes.
  • Thus the present invention makes it possible to prevent and/or treat psoriasis or reduce some of its effects.
  • 3.1.5) Eczema
  • Eczema is a skin complaint characterized clinically by blotches, more or less extensive, localized swellings, and weeping vesicles that subsequently form scabs, accompanied by intense itching. In its chronic phase eczema is complicated by a modification of the skin with thickening. Apoptosis is reduced in the epidermis.
  • The inventors identified a very strong expression of LOX located at the periphery of the cells. On the other hand, NRAGE is weakly expressed and is observed only in the cytoplasm, which certainly reflects a non-existent or very weak level of interaction between the two proteins studied.
  • Thus stimulation of the expression (and/or activity) of NRAGE, and/or inhibition, preferably partial inhibition, of the expression (and/or activity) of LOX, makes it possible to increase apoptosis and thereby to reduce some of the effects of eczema.
  • Thus the present invention makes it possible to prevent and/or treat eczema and in particular to reduce some of its effects.
  • 3.1.6) Epithelial Skin Cancers
  • A distinction is made between two major types of epithelial skin carcinoma:
  • basocellular carcinoma (90% of cases) is an essentially local, slowly developing tumor which virtually never metastasizes. It results from an uncontrolled proliferation of the keratinocytes of the basal cell layer;
  • spinocellular carcinoma (10% of cases) has a much more aggressive local development and can metastasize. It originates from an uncontrolled proliferation of the keratinocytes of the prickle cell layer.
  • The inventors identified the absence of LOX and NRAGE in the invasive cells of the two types of cancer studied (basocellular and spinocellular cancers), with a progressive loss of LOX and NRAGE in the epidermis in the vicinity of the tumors. The inventors also observed that LOX is strongly expressed in the stromal reaction around the tumors, whereas NRAGE is absent in this reaction.
  • It may be noted at this stage that the loss of expression of LOX, which had never previously been identified in the case of these two types of cancer, is unexpected and is perhaps specific to epithelial cancers, since LOX is generally considered to be present in cancers in situ.
  • The loss of expression of NRAGE is completely novel as well and has never been described in any cancer.
  • Thus stimulation of the expression (and/or activity) of NRAGE and the expression (and/or activity) of LOX makes it possible to restore homeostasis. It is possible to bring about a reversion of the tumoral phenotype, especially in epithelial skin cancers.
  • 3.2) Conclusion on the Studies on Pathological Tissues
  • On the basis of these observations the inventors were able to demonstrate that the situations involving a deregulation of the balance between proliferation, differentiation and apoptosis, whether this be in the epidermis of elderly subjects or subjects suffering from different diseases affecting the epidermis in particular, were systematically characterized by defects in the expression of LOX and/or NRAGE, said defects being likely to change their interaction or render it impossible.
  • This finding led the inventors to search for a means of restoring the balance between proliferation, differentiation and apoptosis by utilizing the control exerted by LOX or NRAGE, preferably by utilizing the LOX-NRAGE couple.
  • Thus, on the basis of these unexpected discoveries, the inventors provided methods of identifying active principles that modulate the expression of LOX and/or NRAGE in order to restore the control exerted by these proteins via their interaction, for the purpose of identifying active principles for the preparation of compositions, especially cosmetic or pharmaceutical compositions.
  • In this way, in the case of epidermal hypoproliferation (aged skin, GVH), which is characterized by an extremely thin epidermis and the absence of expression of LOX, the expression of LOX is stimulated, with or without inhibiting the expression of NRAGE, in order to reinduce, via modulation of the co-expression of LOX and NRAGE, a regulated differentiation and apoptosis zone that leads to a thickening of the skin.
  • In the case of epidermal hyperproliferation (psoriasis, eczema), where NRAGE is underexpressed, the expression of NRAGE is stimulated, with or without (slightly) inhibiting that of LOX, in order to promote apoptosis by reinducing a regulated proliferation zone (LOX/NRAGE overlap) and making it possible to stop the hyperproliferation.
  • In the case of a high level of apoptosis in the epidermis (aged skin, exposure of the skin to a stress, particularly exposure to heat, or exposure of the skin to radiation, particularly solar radiation, or exposure of the skin to a toxic agent, for example a chemical or microbiological agent, or graft-versus-host reaction—GVH), where NRAGE is overexpressed, the expression of NRAGE is inhibited, with or without stimulating the expression of LOX.
  • 4) Identification of the Involvement of LOX in Cellular Apoptosis
  • Unexpectedly, the inventors identified the existence of a hitherto unknown link between LOX and apoptosis. Thus the data obtained reflect the antiapoptotic role of LOX in the keratinocytes, said role involving regulation of the proapoptotic protein NRAGE in particular.
  • 5) Search for Active Principles
  • Active principles were identified in particular by analyzing the expression of the messenger RNAs of LOX and NRAGE, especially on keratinocytes in culture and preferably on human keratinocytes. The active principles whose activity is to be tested are placed in contact with the keratinocytes in culture for a sufficient time and under appropriate conditions for the contact to be effective. The active principles are preferably tested in different concentrations so that any influence of concentration can be detected.
  • Advantageously, the active principles screened in the present invention are of vegetable origin, in particular so as to avoid the problems associated with chemical synthesis. The advantages of active principles of vegetable origin are well known to those skilled in the art, especially in pharmacy, dermopharmacy, neutraceutics and cosmetics.
  • The search for active principles is carried out especially by extracting the total RNAs and then performing quantitative RT-PCR. In particular, the preferred primer sequences are those used in Example 13, without implying a limitation.
  • The amount of cDNA in each assay is plotted against the amount of actin cDNA. The effect of the presence or absence of active principles is then compared. If the expression and/or activity of NRAGE and/or LOX are modulated (stimulated or inhibited) relative to the controls, the substance can then be qualified as an active principle. Advantageously, an active principle makes it possible to modulate the expression of the messenger RNA of LOX and/or NRAGE by at least 50% or the expression and/or activity of LOX and/or NRAGE by at least 15%.
  • The compounds according to the present invention are prepared in the form of compositions, especially cosmetic, dermopharmaceutical or pharmaceutical compositions. The excipient for these compositions therefore contains e.g. at least one compound selected from the group consisting of preservatives, emollients, emulsifiers, surfactants, moisturizers, thickeners, conditioners, matting agents, stabilizers, antioxidants, texturizing agents, brightening agents, film-forming agents, solubilizers, pigments, colorants, perfumes and sun filters. These excipients are preferably selected from the group consisting of amino acids and their derivatives, polyglycerols, esters, cellulose polymers and derivatives, lanolin derivatives, phospholipids, lactoferrins, lactoperoxidases, sucrose-based stabilizers, vitamin E and its derivatives, natural and synthetic waxes, vegetable oils, triglycerides, unsaponifiables, phytosterols, plant esters, silicones and their derivatives, protein hydrolyzates, jojoba oil and its derivatives, liposoluble/water-soluble esters, betaines, amine oxides, plant extracts, sucrose esters, titanium dioxides, glycines and parabens, and particularly preferably from the group consisting of butylene glycol, steareth-2, steareth-21, glycol-15 stearyl ether, cetearyl alcohol, phenoxyethanol, methylparaben, ethylparaben, propylparaben, butylparaben, butylene glycol, natural tocopherols, glycerol, sodium dihydroxycetyl phosphate, isopropyl hydroxycetyl ether, glycol stearate, triisononanoin, octyl cocoate, polyacrylamide, isoparaffin, laureth-7, a carbomer, propylene glycol, glycerol, bisabolol, a dimethicone, sodium hydroxide, PEG-30 dipolyhydroxystearate, capric/caprylic triglycerides, cetearyl octanoate, dibutyl adipate, grapeseed oil, jojoba oil, magnesium sulfate, EDTA, a cyclomethicone, xanthan gum, citric acid, sodium laurylsulfate, mineral waxes and oils, isostearyl isostearate, propylene glycol dipelargonate, propylene glycol isostearate, PEG-8 beeswax, hydrogenated palm kernel glycerides, hydrogenated palm glycerides, lanolin oil, sesame oil, cetyl lactate, lanolin alcohol, castor oil, titanium dioxide, lactose, sucrose, low density polyethylene and an isotonic saline solution.
  • Advantageously, the abovementioned compositions are formulated in a form selected from the group consisting of an aqueous or oily solution, a cream or an aqueous or oily gel, particularly in a pot or tube, especially a shower gel or a shampoo; a milk; an emulsion, microemulsion or nanoemulsion, particularly an oil-in-water, water-in-oil, multiple or silicone emulsion; a lotion, particularly in a glass or plastic bottle, a dosing bottle or an aerosol; an ampoule; a syrup; a liquid soap; a hypoallergenic cleansing bar; an ointment; a foam; an injectable solution; an anhydrous, preferably liquid, pasty or solid product, for example in stick form, especially in the form of lipstick; a powder; and a tablet.
  • In the Figures:
  • FIG. 1 shows a sequence and a diagram of the NRAGE protein;
  • FIG. 2 shows the mean of the results obtained in Example 2 for the two-hybrid interaction;
  • FIG. 3 shows the identification of the presence of LOX and NRAGE in reconstructed skin;
  • FIG. 4 shows the location of LOX and NRAGE on human skin sections by confocal microscopy;
  • FIG. 5 shows the detection of LOX and NRAGE on' human skin sections from a 91-year-old donor;
  • FIG. 6 shows the detection of LOX and NRAGE in the skin of a person suffering from graft-versus-host reaction;
  • FIG. 7 shows the detection of LOX and NRAGE in the skin of a patient presenting with a cancer of basocellular or spinocellular type;
  • FIG. 8 shows the detection of LOX and NRAGE in the skin of a patient suffering from lichen planus;
  • FIG. 9 shows the identification of the location of LOX and NRAGE in the skin of a patient suffering from psoriasis;
  • FIG. 10 shows the identification of the location of LOX and NRAGE in the skin of a patient suffering from eczema.
  • FIG. 11 shows the global labelling of the reconstructed skin with Evans blue (red epidermal layers, blue cells, red fibers of the dermal substrate, dermo-epidermal junction in dotted lines).
  • FIG. 12 shows the immuno-histochemical detection of cyto-keratin 10 on the reconstructed skin (cells strongly expressing the red label, the nuclei in blue, the dermo-epidermal junction in dotted lines).
  • FIG. 13 shows the immuno-histochemical detection of transglutaminase on the reconstructed skin (cells strongly expressing the red label, the nuclei in blue, the dermo-epidermal junction in dotted lines).
  • Other objects, characteristics and advantages of the invention will become clearly apparent to those skilled in the art from the explanatory description referring to Examples, which are given solely by way of illustration and cannot in any way limit the scope of the invention.
  • The Examples form an integral part of the present invention and any characteristic that appears novel relative to any prior art on the basis of the description taken in its entirety, including the Examples, forms an integral part of the invention in its function and in its general applicability.
  • Each Example thus has a general scope.
  • Furthermore, in the Examples, unless stated otherwise, all the percentages are given by weight, the temperature is expressed in degrees Celsius and the pressure is atmospheric pressure.
  • EXAMPLES Example 1 Cloning of NRAGE by the Yeast Two-Hybrid Technique
  • The invention initially consisted in searching for potential partners for LOX in keratinocytes. The yeast two-hybrid technique (Y2H) was employed. The Y2H system makes it possible to identify and characterize interactions between a “lure” protein and “target” potential partners. In the present case, the lure was the mature region of LOX fused to the DNA binding domain (BD) of the Ga14 gene. The target or targets were the genes encoded by a complementary DNA library of human keratinocytes, fused to the activation domain (AD) of the Gal4 gene.
  • Interaction of the domains of the lure (LOX) with the domains encoded by a library sequence allows binding and activation of the Gal4 promoter, which controls different genes that confer auxotrophy to AH109 yeasts, enabling growth on deficient medium and activation of the galactosidase activities. A gene coding for a protein that was a candidate partner for LOX was thus identified by virtue of this technique for the possible selection of interactants: the intracellular protein Melanoma Associated Antigen D1 (MAGE-D1) or NRAGE (FIG. 1).
  • Screening of a Library of Keratinocytes by the Yeast Two-Hybrid Technique Using the Lure GAL4 BD-hLOXmat (Human LOX Mature)
  • The cDNA library of normal human skin keratinocytes in vector pGAD-10 was used. The lure chosen for this screening was LOXmat encoded by the human cDNA region of the LOX enzyme without the signal peptide or the pro-region. The nucleotide sequence was inserted in phase behind the DNA binding domain (BD) of the GAL4 transcription factor in vector pBD-Gal4 Cam. The LOX fragment +494 to +1254 to be inserted was amplified by PCR (polymerase chain reaction), and inserted behind the Gal4 binding domain in vector pGal4-BD, using the following primers:
  • 1) (introducing Bam HI)
    ggg atc cgc atg gtg ggc gac gac
    2) (introducing Sal I at the stop which
    is conserved)
    ttg tcg act aat acg gtg aaa ttg tgc.
  • The amplified fragment was initially introduced into vector TOPO and then inserted into pGa14-BD. To validate the expression of the BD-LOXmat fusion protein, the AH109 yeasts were transformed by lure plasmid pBD-LOXmat.
  • The transformed yeasts are screened: 6.88×106 cfu (growth in deficient medium) in the AH109 yeast, 80 selected clones growing in adenine, histidine, tryptophan and leucine deficient medium, 23 of which were retained for their strong growth.
  • Example 2 Validation of the Interaction Between LOX and NRAGE by the Mammalian Two-Hybrid Technique (M2H)
  • Hela cells are transfected on the one hand by plasmids pAct and pAct-NRAGE and on the other hand by pBind and pBind-LOXmat in the presence of lipofectamine. The luciferase activity is tested after 48 h and the results are expressed as the ratio of luciferase activity to control (empty pBind vector). The experiments are performed in triplicate; the mean of the results obtained is shown in FIG. 2.
  • The hLOX sequence used in Example 1 for yeast two-hybrid screening was inserted in phase behind the Gal4 binding domain in the pBind vector (Promega, Madison, USA) for the mammalian two-hybrid interactions.
  • The NRAGE prey was inserted behind the VP16 Gal4 activation domain in the pAct vector (Promega). It starts at amino acid 152 (nucleotide 458).
  • Example 3 Co-Immunoprecipitation of LOX and NRAGE in Mammalian Cells
  • Cos7 cells (mammalian kidney epithelial cells) are co-transfected for the LOX genes (human complete, human mature region and murine mature region) by the construct pcLOX32-V5His (LOX), pcLOX36-V5His or pcLOX27-V5His and for the NRAGE gene by pNM3-HA (complete NRAGE) or pNM7-HA (IRD region). The transfection is performed in Petri dishes 100 mm in diameter using lipofectamine. After 48 h the transfected cells are lyzed in 500 μl of lysis buffer. The proteins of the cell lyzates are incubated with either anti-V5 or anti-HA; the anti-V5 or anti-HA monoclonal antibody is added to the cell lyzate at a rate of 1/250 over 1 h 30 min at 4° C., with shaking.
  • The immune complexes (ICs) formed in this way are precipitated with protein G-Sepharose over 1 h at 4° C., with shaking. After three 10-minute washes in lysis buffer and elution in SDS-PAGE buffer, all the ICs are subjected to electrophoresis on a 10% SDS-PAGE gel, followed by Western blotting, Transfer of the ICs to a PVDF (polyvinylidene fluoride) membrane is followed by development:
  • for NRAGE, with anti-HA diluted to 1/1000, then anti-mouse-HRP (horse radish peroxidase) diluted to 1/20,000,
  • for LOX, with anti-VS-HRP diluted to 1/5000.
  • The final detection is effected by chemoluminescence of the HRP.
  • The results obtained demonstrate that the complete form of NRAGE (NM3-HA) co-immunoprecipitates with the human complete form of LOX (LOX 32H), mature human LOX (LOX 36H) and mature murine LOX (LOX 27H). Furthermore, NRAGE (NM7-HA), which corresponds to the IRD region, co-immunoprecipitates in the same way with these three recombinant proteins, showing the involvement of this region in the interaction.
  • Example 4 Identification, by Immunohistochemistry, of the Presence of LOX and NRAGE in a Model of Reconstructed Skin
  • The identification was effected in a model of reconstructed skin (MIMESKIN®, Engelhard Lyon, France) prepared from a dermal substrate (collagen/glycosaminoglycans/chitosan, MIMEDISC®, Engelhard Lyon, France) inoculated with normal human fibroblasts, on whose surface normal human keratinocytes were deposited.
  • After 45 days of culture, allowing differentiation of the keratinocytes by exposure at the air-liquid interface, the samples are fixed in Bouin's fixative or formaldehyde and then included in paraffin.
  • Sections are immunolabeled with the antibodies described below:
  • anti-LOX antibody obtained and purified by the method described by Sommer et al. (Transient expression of lysyl oxidase by liver myofibroblasts in murine schistosomiasis, Laboratory Investigation, 69, 460-470, 1993),
  • anti-NRAGE antibody (goat polyclonal IgG),
  • rabbit anti-goat secondary antibody.
  • The immune complexes are detected with a peroxidase-conjugated anti-rabbit IgG using diaminobenzidine as substrate, followed by counterstaining with hematoxylin.
  • FIG. 3 shows the immunohistochemical detection of LOX and NRAGE on the reconstructed skin.
  • The position of the dermo-epidermal junction is indicated by a continuous line, the position of the dermal substrate is indicated by an arrow and the location of the keratinocytes is indicated by an arrowhead.
  • The expression of LOX appears from the basal cell layer upwards, persists in the suprabasal cell layers and progressively disappears in the differentiated layers. The expression of NRAGE is slightly shifted; it asserts itself from the non-proliferative suprabasal cell layers upwards and intensifies greatly in the granular and horny layers, where LOX is no longer expressed.
  • These proteins are therefore associated in the non-proliferative suprabasal cell layers of the epidermis of the MIMESKIKN® model of reconstructed skin. This co-location was confirmed by immunohistology (IH) on normal human skin.
  • The inventors thus demonstrated that the LOX and NRAGE proteins are expressed in the epidermis, with a co-location zone in the prickle cell layers.
  • Example 5 Identification of the Co-Location of LOX and NRAGE in Normal Human Skin by Confocal Microscopy
  • Frozen sections of samples of normal human skin originating from surgical resection (foreskin) are prepared. The anti-LOX and anti-NRAGE primary antibodies of Example 4 were used to detect the expression of LOX and NRAGE. The secondary antibodies used are:
  • donkey anti-rabbit IgG-F1TC, green fluorescein labeling,
  • donkey anti-goat IgG-R, red rhodamine labeling.
  • A negative control was prepared in the absence of primary antibodies.
  • The dual labeling was observed using a ZEISS AXIOPLAN 2 LSM510 confocal upright microscope and the images were acquired using ZEISS LSM5 Image Browser software.
  • FIG. 4 shows the immunodetection of LOX and NRAGE in normal human skin by confocal microscopy.
  • The observations confirm the results of Example 4, further illustrating the co-location of LOX and NRAGE in the prickle cell layers of the epidermis of no mal human skin, with an almost perfect juxtaposition of the expression of LOX and NRAGE. Thus the observations at the cellular level show that LOX and NRAGE appear at the periphery of the cell (in the submembranous peripheral zone), NRAGE also appearing in the cytoplasm.
  • The inventors thus demonstrated fulfilment of the conditions for the direct interaction they had identified in vitro to be able to take place in the epidermis in the zone where both LOX and NRAGE are located.
  • Example 6 Identification of the Location of LOX and NRAGE in the Skin of Persons of Different Ages
  • An immunohistological study using the protocol described in Example 4 was carried out on sections of human skin originating from 2 donors from different age groups (under 20 years and over 60 years).
  • FIG. 5 shows the labeling performed on the skin of a 91-year-old donor. The observations show a hypoproliferative epidermis that is very thin (reduced to a few cell layers) and hyperkeratinized.
  • The inventors identified the total absence of LOX (detectable by the technique used). On the other hand, NRAGE is strongly expressed, is located in the cytoplasm and appears from the first suprabasal cell layer upwards, without an expression gradient.
  • Example 7 Detection of LOX and NRAGE in the Skin of a Person Suffering From Graft-Versus-Host Reaction (GVH)
  • An immunohistological study using the protocol described in Example 4 was carried out on sections of human skin from patients suffering from graft-versus-host reaction (GVH). This study involved 5 different donors.
  • FIG. 6 identifies an almost total disappearance of LOX in the epidermis (without modification of its expression in the dermis) and a very pronounced presence of NRAGE, which is located in the cytoplasm and appears from the first suprabasal cell layers upwards.
  • Example 8 Detection of LOX and NRAGE in the Skin of a Person Presenting with a Cancer of Basocellular and Spinocellular Type
  • An immunohistological study using the protocol described in Example 4 was carried out on samples of skin from patients presenting with a basocellular or spinocellular cancer.
  • FIG. 7 shows the following in both types of cancer studied:
  • a progressive decrease in the expression of LOX and NRAGE in the epidermis at the periphery of the tumors,
  • the absence of LOX and NRAGE in the epidermal invasive cells,
  • a strong expression of LOX in the dermal stromal reaction around the tumors,
  • the absence of NRAGE in the dermal stromal reaction around the tumors.
  • Example 9 Detection of LOX and NRAGE in the Skin of a Patient Suffering From Lichen Planus
  • An immunohistological study using the protocol described in Example 4 was carried out on sections of skin from 3 patients suffering from lichen planus.
  • FIG. 8 shows a very large decrease in the expression of LOX in the epidermis, or even its total absence. This disturbance correlates with an irregularity in the expression of NRAGE in the epidermis.
  • Example 10 Identification of the Location of LOX and NRAGE in the Skin of a Patient Suffering From Psoriasis
  • An immunohistological study using the protocol described in Example 4 was carried out on sections of skin from 5 patients suffering from psoriasis.
  • FIG. 9 is representative of all the sections observed and shows a very strong expression of LOX and a moderate presence of NRAGE in the epidermis, with a more or less homogeneous labeling of the zones in question, showing no expression gradient. These characteristics of an intensity of expression varying from the norm are coupled with much more important anomalies affecting the location of the proteins in question. Thus, in psoriatic skin, LOX is expressed essentially in the lower part of the epidermis and NRAGE solely in its upper part, the expression of the proteins thus being shifted without the overlap zone normally observed in healthy skin. At the cellular level, NRAGE is observed only in the cytoplasm and not in the submembranous peripheral zone.
  • Example 11 Identification of the Location of LOX and NRAGE in the Skin of a Patient Suffering From Eczema
  • An immunohistological study using the protocol described in Example 4 was carried out on sections of skin from patients suffering from eczema.
  • FIG. 10 shows a very strong pericellular expression of LOX in the epidermis. On the other hand, NRAGE is weakly expressed and is observed only in the cytoplasm, which reflects a loss of co-location at the cellular level.
  • Example 12 Identification of the Involvement of LOX in the Inhibition of Apoptosis
  • The study was carried out on monolayer cultures of differentiated human keratinocytes at confluence. The effect of LOX on apoptosis was identified by inhibiting its activity through the addition of β-APN (0.02% w/v—(weight/volume)) under normal or proapoptotic conditions (thermal shock caused by exposure to a temperature of 45° C.±0.5° C. for 1 h 30 min).
  • Cell death by apoptosis is detected and quantified using the TUNEL (terminal deoxynucleotidyl transferase mediated dUTP nick end labeling) technique, which is based on labeling of the DNA deletions accompanying apoptosis.
  • The first step consists in labeling the DNA breaks with TdT (terminal deoxynucleotidyl transferase), which catalyzes the polymerization of the fluorescein-labeled nucleotides at the free 3′-OH end of the DNA. The second step consists in detecting the incorporated fluorescein with an alkaline phosphatase-conjugated anti-fluorescein antibody after incubation with the substrate for the latter.
  • The experiment is validated by means of a positive control obtained by fragmentation of the DNA with a DNase solution, and a negative control obtained by depositing phosphate buffer.
  • The results obtained are shown in the Table below.
  • TABLE 1
    Without thermal shock With thermal shock
    (% of labeled cells) (% of labeled cells)
    Without β-APN  6% 39%
    With β-APN 56% 81%
  • The results obtained demonstrate that:
  • thermal shock (45° C.±0.5° C. for 1 h 30 min) induces pronounced cellular apoptosis, inhibition of the enzymatic activity of LOX by β-APN causes a substantial increase in apoptosis.
  • These results identify an antiapoptotic effect of the activity of LOX.
  • Example 13 Analysis of the Expression of LOX and NRAGE Messenger RNAs by Differentiated Keratinocytes in Culture in a Calcium Medium, With and Without Contact With Active Principles Whose Activity is to be Tested (Analysis Performed e.g. by Quantitative RT-PCR, Screening of Active Principles)
  • The active principles were tested on keratinocytes of normal human foreskins from young subjects (pooled normal human foreskin epidermal keratinocytes—Clonetics).
  • The keratinocytes are amplified e.g. in K-SFM (keratinocyte serum-free medium), supplemented with antibiotics, up to the third passage at 37° C. under 5% CO2.
  • The cells are inoculated into 96-well plates, e.g. at a rate of 40,000 cells per cm2, and cultivated to about 80% confluence. The cells are then cultivated in hypercalcium medium (1.7 mM CaCl2 at 37° C. under 5% CO2) to induce differentiation of the cells.
  • The starting material used to prepare the active principles, in the case of plants (preferably roots, stems, barks, flowers, fruits, seeds, germs, gums, exudates, leaves or whole plants) or proteins, may or may not be sterilized by radiation, for example beta or gamma radiation preferably at a dose of 5 kGy, and is then reduced to powder if necessary, for example by grinding at room temperature. The powder is then dispersed at a rate of 2 to 5% (weight/weight) of powder, preferably 5%, either in a polar solvent, e.g. water or butylene glycol, and/or a mixture of polar solvents, advantageously a mixture of water and an alcohol, glycol or polyol (such as ethanol, glycerol, butylene glycol and other glycols, xylitol, etc.) in variable proportions, and preferably a 75/25 or 50/50 water/butylene glycol mixture, or in an apolar solvent, e.g. an alkane, or a mixture of apolar solvents, or in a mixture of polar and apolar solvents. After stirring, e.g. magnetic stirring, for at least 2 hours, the sample is clarified by decantation or centrifugation and then filtered, preferably on a 0.45 μm or 0.22 μm filter.
  • The starting material used to prepare the active principles, in the case of characterized molecules (e.g. molecules obtained by synthesis or hemisynthesis, biological molecules obtained by purification), is diluted in a solvent, preferably water or dimethyl sulfoxide (in a concentration preferably of between 10−6 M and 10−2 M and particularly preferably in the order of 10−4 M, or preferably of between 1% weight/weight and 5% weight/weight, depending on the molecules). The solution obtained is then optionally filtered, preferably on a 0.45 μm or 0.22 μm filter.
  • The active principles obtained by one of the methods described above are then tested in a final concentration preferably of between 0.01% volume/volume (v/v) and 10% (v/v) and advantageously of between 0.1% and 1% (v/v), for example at 1% (v/v).
  • Incubation in the presence of the cells is advantageously carried out for 24 hours in hypercalcium K-SFM without growth factors. The cells are frozen dry at −80° C. after being rinsed in phosphate buffer pH 7.4.
  • Extraction of the Total RNAs
  • The total RNAs are extracted with the SV Total RNA Isolation System (Promega, Meylan, France) for the conditions of 96-well plates according to the manufacturer's protocol.
  • The gene expression is modified by real-time RT-PCR, measuring the expression of each gene relative to actin (housekeeping gene) and expressed in % of the untreated negative control.
  • Quantitative real-time RT-PCR (Q-RT-PCR)
  • 10 μl of total RNAs at 5 ng/μl are added to 40 μl of PCR mix (composed of 25 μl of SYBR Green Buffer Mix 2×, 0.5 μl of enzyme mix, a final concentration of 0.5 μM of sense primer and a final concentration of 0.5 μM of antisense primer, and RNase and DNase free water qsp 40 μl).
  • The RT-PCR proceeds in different steps comprising retrotranscription at 50° C., 30 min, polymerase activation at 95° C., 15 min, and execution of the PCR cycles (95° C., 15 s; 60° C., 30 s; 72° C., 30 s)×50 cycles.
  • Production of the Fusion Curve
  • 90° C., 1 min
  • 30° C., 1 min
  • 50° C. to 95° C., 10 s/° C. (fusion curve)
  • The percentage stimulation or inhibition is expressed relative to the untreated control (in the absence of test substance).
  • Actin gene-Hybridization 60° C.
    Sense
    GTGGGGCGCCCCAGGCACCA
    Antisense
    CTCCTTAATGTCACGCACGATTTC
    LOX gene-Hybridization 60° C.
    Sense
    ACGTACGTGCAGAAGATGTCC
    Antisense
    GGCTGGGTAAGAAATCTGATG
    NRAGE gene-Hybridization 60° C.
    Sense
    TGCACAGACATCAGCAGATGG
    Antisense
    TTCACGGATGATATCTCTCAGC
    Involucrin gene-Hybridization 60° C.
    Sense
    TGTTCCTCCTCCAGTCAATACCC
    Antisense
    ATTCCTCATGCTGTTCCCAGTGC
  • To take account of the cell population present, all the results were compared with the “actin” signal used as a housekeeping gene. Depending on the experiment, the measurement threshold of the C(T) (=cycle threshold) was fixed for a value of T of between 0.05 and 0.01, an arbitrary unit of measurement then being calculated for each gene according to the following formula:

  • Sgene <<x>> 107×(1/2)C(T)gene <<x>>
  • C(T)gene <<x>> denotes the number of cycles necessary to reach the fluorescence threshold of 0.01-0.05 of the gene <<x>>.
  • The values of the genes of interest were compared with the “actin” signal by calculation of the following ratio:

  • R=Sgene<<x>>/Sactin
  • These ratios were compared between the treated and untreated samples, <<x>> being the actin, LOX or NRAGE gene.
  • Screening of Active Principles
  • The amounts of cDNA in each assay are compared with the amount of actin cDNA and then with the negative controls (without active principles). The results are considered significant if the measured effect reaches a factor of about 2. Of 120 active principles tested, 3 correspond to these criteria at the concentrations tested and under the conditions defined. These active principles form the subject of the Table below:
  • TABLE 2
    NRAGE LOX
    Name Control multiplied by: Control multiplied by:
    Ephedra 2 2
    Soya 2.5 1
    Hops 1 2
  • Preferably, the ephedra extract is obtained by extraction of the whole plant, especially with a polar solvent such as water or a water/butylene glycol mixture (e.g. 75/25 or 50/50), preferably water.
  • Preferably, the hop extract is obtained by extraction of the cones, especially with a polar solvent such as water or a water/butylene glycol mixture (75/25 or 50/50), preferably water.
  • Preferably, the soya extract is obtained by extraction of the seed, especially with a polar solvent such as water or a water/butylene glycol mixture (75/25 or 50/50), preferably water.
  • Conclusions
  • Of the bank of 120 active principles under the conditions considered:
  • 1 active principle is capable of significantly activating the rate of synthesis of mRNA of the genes coding for NRAGE and LOX,
  • 1 active principle is capable of significantly activating the rate of synthesis of mRNA of the gene coding for NRAGE, without having an effect on the gene coding for LOX,
  • and 1 active principle is capable of significantly activating the rate of synthesis of mRNA of the gene coding for LOX, without having an effect on the gene coding for NRAGE.
  • This study made it possible to select active principles capable of modulating the balance between proliferation, differentiation and apoptosis, at least in the keratinocytes.
  • The ephedra extract can be used e.g. in the treatment of diseases such as cancers, preferably cutaneous epithelial cancers (basocellular or spinocellular), or lichen planus, or optionally in the treatment of certain cutaneous manifestations of GVH, or else to reduce the effects of ageing on the skin.
  • The soya extract can be used e.g. in the treatment of diseases such as cancers, like cutaneous epithelial cancers (basocellular or spinocellular), GVH or lichen planus, or for combating or preventing ageing. The soya extract can be used especially for combating cell hyperproliferation.
  • The hop extract can be used e.g. in the treatment of diseases such as cancers, like cutaneous epithelial cancers (basocellular or spinocellular), eczema or psoriasis. The hop extract can be used especially for combating cell hypo-proliferation.
  • The soya extract and hop extract can be used in association e.g. in the treatment of diseases such as cancers, like cutaneous epithelial cancers (basocellular or spinocellular), or lichen planus.
  • The soya extract and ephedra extract can be used in association e.g. in the treatment of diseases such as cancers, like cutaneous epithelial cancers (basocellular or spinocellular), or lichen planus.
  • The hop extract and ephedra extract can be used in association e.g. in the treatment of diseases such as cancers, like cutaneous epithelial cancers (basocellular or spinocellular), or lichen planus.
  • Example 14 Analysis, by Quantitative RT-PCR, of the Kinetics of Expression of the Messenger RNAs of NRAGE and LOX on Keratinocytes During Calcium Differentiation, and Effect of Active Principles on the Expression Kinetics
  • The experimental conditions used to obtain cells at 80% confluence are identical to those described in Example 13. Differentiation takes place in the presence of calcium (1.7 mM CaCl2) and active principle. An analysis is performed by Q-RT-PCR (method described in Example 13) after 2, 3 and 4 days of incubation.
  • Nine other substances were tested in this way. One of them, a cinnamon extract, affords an inhibition of LOX and NRAGE under the experimental conditions considered.
  • TABLE 3
    NRAGE LOX
    Name Control multiplied by Control multiplied by
    Cinnamon
    2 D 0.9 0.5
    3 D 0.7 0.7
    4 D 0.2 0.5
    (D = Days)
  • Preferably, the cinnamon extract is obtained by extraction of the bark, especially with a polar solvent such as water or a water/butylene glycol mixture (75/25 or 50/50), preferably water.
  • Conclusion
  • The cinnamon extract can be used e.g. in the treatment of diseases such as psoriasis.
  • The cinnamon extract and soya extract (whose effect was detected in the previous Example) can be used in association e.g. in the treatment of diseases such as certain cutaneous manifestations of GVH, eczema or psoriasis, or else for reducing the effects of ageing on the skin.
  • Example 15 Analysis of the Expression of LOX Messenger RNAs by Keratinocytes in Culture in the Absence of Calcium Differentiation, With and Without Their Being Put Into Contact With the Active Ingredients, the Activity of Which is to be Tested (an Analysis For Example, Achieved by Quantitative RT-PCR, Screening of Active Ingredients)
  • The actives were tested on normal human keratinocytes from young subjects, obtained by enzymatic extraction of human biopsies collected after surgical resection and cultivated as monolayers in a K-SFM (keratinocyte serum free medium with supplements) defined medium, supplemented with antibiotics at 37° C. under 5% CO2.
  • The cells are sown at the second passage in 24-well plates, for example with an amount of 30 000 cells per cm2 and grown until about 95% confluence. The cell carpets are rinsed with phosphate buffer at pH 7.4 preferably with calcium and magnesium, before being put into contact with the active ingredients to be tested or the reference positive controls diluted in the K-SFM medium, prepared without supplements but with antibiotics.
  • Active ingredients of different origins (vegetable, biotechnological origin or synthesized molecules for example) are tested from 0.1% volume/volume (v/v) to 1% (v/v). An active ingredient from vegetable origin is tested for example at 1% (v/v) and a synthesized molecule is tested for example at 0.1% (v/v)
  • In particular, the active ingredient from vegetable origin are extracts which are obtained by macerating plants (preferably roots, rhizomes, stems, barks, flowers, fruit, seeds, germs or leaves) at 2-5% (w/w) in a solvent or a mixture of solvents, advantageously a 100:0 to 0:100 water/(alcohol, glycol or polyol) (such as ethanol, glycerol, butylene glycol and other glycols, xylitol, etc . . . ) mixture. The obtained extracts are then filtered or distilled in order to recover the soluble fraction which is then filtered at 0.45 μm preferably. The biotechnological hydrolysates are obtained by fermentation of vegetable extracts in the presence of microorganisms, advantageously from the Lactobacillus or Saccharomyces family. These hydrolysates are then preferably filtered to 0.45 μm.
  • The incubation is advantageously performed for 24 hours at 37° C. under 5% CO2, in a K-SFM medium without growth factors but with antibiotics. The negative controls are either the culture medium by itself, or the culture medium containing from 0.1% (v/v) to 1% (v/v) of the solvent used during the method for extracting the tested extracts. The reference positive control used for inducing cell differentiation is a solution of calcium chloride (CaCl2 - final concentration of 1.7 mM).
  • The untreated cells (NT control) are dry frozen at −80° C. after rinsing with pH 7.4 phosphate buffer. After treatment for 24 hrs in the presence of actives or controls, the cells are dry frozen at −80° C. after rinsing with pH 7.4 phosphate buffer.
  • Extraction of Total RNAs
  • The total RNAs are extracted by means of the SV total RNA isolation system (Promega, Meylan, France) for the conditions of 24-well plates according to the protocol of the manufacturer.
  • Modification of the expression of the genes is achieved by real time RT-PCR which measures the expression of each gene relative to actine (housekeeping gene) and expressed as a % of the untreated negative control (NT).
  • Quantitative Real Time RT-PCR (Q-RT-PCR)
  • 10 μl of 5 ng/μl total RNAs are added to 40 μl of PCR mix (consisting of 25 μl of SYBR Green Buffer Mix 2×, 0.5 μl of enzyme mix, final 0.5 μM of sense primer and final 0.5 μM of antisense primer, RNAse-free and DNase-free water qsp 40 μl).
  • RT-PCR is performed in different steps including retrotranscription at 50° C., 30 min, activation of polymerase at 95° C., 15 min, performing PCR cycles, (95° C.-15 s, 60° C.-30s, 72° C.-63s, 78° C.-30s)×50 cycles.
  • Producing the Melting Curve
  • 90° C., 1 min
  • 30° C., 1 min
  • 50° C. to 95° C., 10 s/° C. (melting curve)
  • Primers Used:
  • Actine gene-hybridization at 60° C.
    Sense
    GTG GGG CGC CCC AGG CAC CA
    Antisense
    CTC CTT AAT GTC ACG CAC GAT TTC
    LOX gene-hybridization at 60° C.
    Sense
    ACG TAC GTG CAG AAG ATG TCC
    Antisense
    GGC TGG GTA AGA AAT CTG ATG
  • In order to take into account the present cell population, all the results are relative to the “actine” signal, used as a housekeeping gene. According to the experimentation, the measuring threshold of C(T) (=cycle threshold) was set for T between 0.05 and 0.01 and then an arbitrary measuring unit is calculated for each gene according to the formula:

  • Sgene “LOX” 107×(1/2) C(T)gene “LOX”
  • C(T)gene “LOX” meaning the number of cycles required for reaching the fluorescence threshold of 0.01-0.05 of the “LOX” gene.
  • The values of the genes of interest were relative to the “actine” signal by calculating the ratio:

  • R=Sgene“LOX”/Sactine.
  • These ratios were compared between the treated and untreated samples.
  • Screening of Active Ingredients:
  • The cDNA amounts of each test are relative to the amount of cDNA of actine and then to the negative controls (NT). The results are considered as significative when the measured effect is a modulation by a factor of about 2 (stimulation) or 0.5 (inhibition). On 60 tested actives, 30 correspond to these criteria under the defined conditions. These actives are the following and are the subject of the Table herein below:
  • TABLE 1
    LOX
    modulation vs Preferred part
    Description Latin name NT of the plant
    Gypsophila Gypsophila 0.4 Root
    Red sandalwood Pterocarpus santalinus 0.4 Total wood
    Common bryony Bryonia dioica 0.5 Root
    Butcher's broom Ruscus aculeatus 2.0 Root
    Lemon Citrus limonia 2.0 Fruit
    Tangerine Citrus reticulate 2.1 Fruit
    Ethyl trans 3-hexanoate 2.3
    Khella Amni Visnaga 2.4 Fruit
    Alginic acid 2.4
    Carrot Daucus Carota 2.4 Root
    Methyl 2 methyl butyrate / 2.5
    Star anise Illicium verum 2.5 Fruit
    Cypress Cupressus sempervirens 2.6 Fruit
    Asae Foetida gum 2.6
    Xylitol / 2.8
    Hops Humulus Lupulus 2.8 Fruit
    Blackthorn Prunus spinosa 3.1 Fruit
    Strawberry tree Arbutus unedo 3.3 Leaf
    Umbellate wintergreen Chimaphila umbellata 3.4 Plant
    Sweet woodruff Asperula odorata 4.7 Plant
    Mugwort Artemisia vulgaris 4.7 Root
    Common elderberry Sambucus nigra 5.0 Fruit
    Chinese cabbage Brassica Brassica campestris 5.3 Plant
    var. Pekinensis
    Cinnamon Cinnamomum spp 5.5 Stem, bark
    Chinese ephedra Ephedra sinica 6.0 Plant
    Bitter ash Cassia amara 6.2 Total wood
    Cacao tree Theobroma cacao 7.4 Fruit shell
    Silk Serica 7.9
    Red sarsaparilla Smilax ornate 9.0 Root
    Redcurrant Ribes rubrum 9.1 Fruit
    Pellitory Annacyclus pyrethrum 9.8 Root
    Sweet fennel Foeniculum 10.2 Plant
  • Conclusions
  • From the bank of 60 actives and under the considered conditions:
      • 3 actives are capable of significatively inhibiting the synthesis rate of mRNA of the LOX coding gene,
      • 28 actives are capable of significatively activating the synthesis rate of mRNA of the LOX coding gene,
  • With this study, it was possible to select active ingredients capable of modulating at least at keratinocyte level, the equilibrium between proliferation, differentiation and apoptosis.
  • Example 15 Analysis of the Expression of Proteins Involved in the Regulation of Epidermal Differentiation/Proliferation Homeostasis in a Reconstructed Skin Model With and Without Contact With Active Ingredients, the Activity of Which is to be Tested by Histology (Example of Ephedra Extract).
  • Demonstration of proliferation or differentiation labels was achieved in a reconstructed skin model (MIMESKIN®, Engelhard Lyons, France) prepared from a dermal substrate (collagen/glycosaminoglycans/chitosan, (MIMEDISK®, Engelhard Lyons, France) sown with normal human fibroblasts, at the surface of which normal human keratinocytes were deposited, the cells being extracted by enzyme treatment from biopsies obtained by surgical resection.
  • The reconstructed skin model is in particular achieved according to the following protocol:
      • 0.5 to 1.106 fibroblasts of normal human skin are sown on a matrix substrate based on collagen/glycosaminoglycan/chitosan, and then grown in a nutritive medium, for example DMEM-Glutamax supplemented with 10% of calf serum; ascorbic acid, preferably at a final concentration of 1 mM; EGF (epidermal grow factor), preferably at a final concentration of 10 ng/ml; Normocin, preferably at a final concentration of 100 μg/ml, for 21 days.
      • 0.5 to 1.106 fibroblasts of normal human skin are sown on the dermal equivalent, and then grown in a nutritive medium, for example DMEM-Glutamax/Ham F-12 (ratio 3:1 v/v) supplemented with calf serum, ascorbic acid, preferably at a final concentration of 1 mM; EGF (epidermal growth factor), preferably at a final concentration of 10 ng/ml; hydrocortisone, preferably at a final concentration of 0.4 μg/ml; umuline, preferably at a final concentration of 0.12 IU/ml; isuprel, preferably at a final concentration of 0.4 μg/mg; triiodothyronine, preferably at a final concentration of 2.10−9M; adenine, preferably at a final concentration of 24.3 μg/ml; Normocin, preferably at a final concentration of 100 μg/ml. The culture was continued for 7 days in an immersed condition. The cultures were then placed at the air-liquid interface for 14 additional days in the same medium as the immersion culture, except for the calf serum, hydrocortisone, isuprel, triidothyronine and umuline.
  • The active ingredient (ephedra extract) is advantageously diluted in the culture media described above at 0.5 and 1% and used 3 days after the respective sowing of fibroblasts and keratinocytes (i.e. days 3 to 21 and days 24 to 42). The positive control is preferably made by adding calcium chloride with a final concentration of 1.5 mM during the emerged phase of the reconstructed skins (i.e. day 28-42) in order to stimulate epidermal differentiation.
  • At the end of the culture, the samples are frozen, included into a thermosensitive resin, and then cryo-cut at 5 μm.
  • The labellings on the cuts were carried tout with the reagents described below:
      • human primary anti-transglutaminase antibody
      • human primary anti-cytokeratin 10 antibody
      • alexa-fluor coupled secondary antibodies
      • Evans blue
      • Dapi
  • Viewing of immunolabellings is performed under photon microscopy (Axioskop2plus—Zeiss, Germany) and quantification of the transglutaminase immunolabellings was performed by image analysis (Lucia—Nilon, France) and the effects of the treatment with the active ingredient are evaluated (Holm-Sidak statistical test, p<0.01).
  • TABLE 1
    Quantification of the labelling intensity of transglutaminase
    INTENSITY Untreated Calcium Ephedra 0.5% Ephedra 1%
    0.149 0.061 0.082 0.276
    0.099 0.076 0.126 0.196
    0.092 0.064 0.094 0.192
    Average 0.113 0.067 0.100 0.221
    Standard deviation 0.031 0.008 0.023 0.048
    Variation vs NT 100%  59%  88% 196%
    Significance vs NT No No Yes
    Variation vs Ca 169% 100% 149% 330%
    Significance vs Ca No No Yes
  • These results show that the active ingredient induces the same type of epidermal differentiation as the positive control with a dose-effect relationship. Indeed, the treated skins have a larger number of keratinocyte layers expressing transglutaminase, in particular in the so-called granular layer. Moreover, the obtained labelling is more intense and more defined as illustrated in the magnification of the epidermal portion. Quantification clearly demonstrates induction of the protein form after treatment, in particular at 1% active ingredient concentration (2-fold induction).
  • FIG. 11 shows the global labelling of the reconstructed skin with Evans blue (red epidermal layers, blue cells, red fibers of the dermal substrate, dermo-epidermal junction in dotted lines).
  • These results show that the active ingredient induces the same type of epidermal differentiation as the positive control with a dose-effect relationship. Indeed, the treated skins have a larger number of differentiated keratinocyte layers and in particular, an increase in the thickness of the so-called granular layer is noted. Moreover, an interesting effect was also obtained on the fibroblast density at the dermas treated with ephedra extract.
  • FIG. 12 shows the immuno-histochemical detection of cyto-keratin 10 on the reconstructed skin (cells strongly expressing the red label, the nuclei in blue, the dermo-epidermal junction in dotted lines).
  • These results show that the active ingredient induces the same type of epidermal differentiation as the positive control with a dose-effect relationship. Indeed, the treated skins have a larger number of keratinocyte layers expressing cytokeratin 10 and in particular in the so-called granular layer.
  • FIG. 13 shows the immuno-histochemical detection of transglutaminase on the reconstructed skin (cells strongly expressing the red label, the nuclei in blue, the dermo-epidermal junction in dotted lines).
  • Example 17 Use of the Products of the Invention in Cosmetic or Pharmaceutical Formulations of the Oil-In-Water Emulsion Type Formulation 17a:
  • A Water qsp 100
    Butylene Glycol 2
    Glycerol 3
    Sodium Dihydroxycetyl 2
    Phosphate,
    Isopropyl Hydroxycetyl Ether
    B Glycol Stearate SE 14
    Triisononanoin 5
    Octyl Cocoate 6
    C Butylene Glycol, Methylparaben, 2
    Ethylparaben, Propylparaben,
    pH adjusted to 5.5
    D Products of the invention 0.01-10%
  • Formulation 17b:
  • A Water qsp 100
    Butylene Glycol 2
    Glycerol 3
    Polyacrylamide, Isoparaffin, 2.8
    Laureth-7
    B Butylene Glycol, Methylparaben, 2
    Ethylparaben, Propylparaben
    Phenoxyethanol, Methylparaben, 2
    Propylparaben, Butylparaben,
    Ethylparaben
    Butylene Glycol 0.5
    D Products of the invention 0.01-10%
  • Formulation 17c:
  • A Carbomer 0.50
    Propylene Glycol 3
    Glycerol 5
    Water qsp 100
    B Octyl Cocoate 5
    Bisabolol 0.30
    Dimethicone 0.30
    C Sodium Hydroxide 1.60
    D Phenoxyethanol, Methylparaben, 0.50
    Propylparaben, Butylparaben,
    Ethylparaben
    E Perfume 0.30
    F Products of the invention 0.01-10%
  • Example 18 of the Invention Use of the Products of the Invention in a Formulation of the Water-In-Oil Type
  • A PEG-30 dipolyhydroxystearate 3
    Capric Triglycerides 3
    Cetearyl Octanoate 4
    Dibutyl Adipate 3
    Grapeseed Oil 1.5
    Jojoba Oil 1.5
    Phenoxyethanol, Methylparaben, 0.5
    Propylparaben, Butylparaben,
    Ethylparaben
    B Glycerol 3
    Butylene Glycol 3
    Magnesium Sulfate 0.5
    EDTA 0.05
    Water qsp 100
    C Cyclomethicone 1
    Dimethicone 1
    D Perfume 0.3
    E Products of the invention 0.01-10%
  • Example 19 of the Invention Use of the Products of the Invention in a Formulation of the Shampoo or Shower Gel Type
  • A Xanthan Gum 0.8
    Water qsp 100
    B Butylene Glycol, Methylparaben, 0.5
    Ethylparaben, Propylparaben
    Phenoxyethanol, Methylparaben, 0.5
    Propylparaben, Butylparaben,
    Ethylparaben
    C Citric acid 0.8
    D Sodium Laureth Sulfate 40.0
    E Product of the invention 0.01-10%
  • Example 20 of the Invention Use of the Products of the Invention in a Formulation of the Lipstick Type and Other Anhydrous Products
  • A Mineral Wax 17.0
    Isostearyl Isostearate 31.5
    Propylene Glycol Dipelargonate 2.6
    Propylene Glycol Isostearate 1.7
    PEG-8 Beeswax 3.0
    Hydrogenated Palm Kernel Oil, 3.4
    Glycerides,
    Hydrogenated Palm Glycerides
    Lanolin Oil 3.4
    Sesame Oil 1.7
    Cetyl Lactate 1.7
    Mineral Oil, Lanolin Alcohol 3.0
    B Castor Oil qsp 100
    Titanium Dioxide 3.9
    CI 15850:1 0.616
    CI 45410:1 0.256
    CI 19140:1 0.048
    CI 77491 2.048
    C Products of the invention 0.01-5%
  • Example 20 of the Invention Use of the Products of the invention in an Aqueous Gel Formulation (Eye Contour Gel, Slimming Gel, Etc.)
  • A Water qsp 100
    Carbomer 0.5
    Butylene Glycol 15
    Phenoxyethanol, Methylparaben, 0.5
    Propylparaben, Butylparaben,
    Ethylparaben
    B Products of the invention 0.01-10%
  • Example 21 of the Invention Use of the Products of the Invention in a Formulation of the Triple Emulsion Type
  • Primary emulsion W1/O
    A PEG-30 dipolyhydroxystearate 4
    Capric Triglycerides 7.5
    Isohexadecane 15
    PPG-15 Stearyl Ether 7.5
    B Water 65.3
    C Phenoxyethanol, Methylparaben, 0.7
    Propylparaben, Butylparaben,
    Ethylparaben
    Secondary emulsion W1/O/W2
    A Primary emulsion 60
    B Poloxamer 407 2
    Phenoxyethanol, Methylparaben, 0.3
    Propylparaben, 2-bromo-2-
    nitropropane-1,3-diol
    Water qsp 100
    C Carbomer 15
    D Triethanolamine pH 6.0-6.5
  • Example 22 of the Invention Preparation of Pharmaceutical Formulations Containing the Product of the Invention Formulation 22a: Preparation of Tablets
  • A Excipients In g per tablet
    Lactose 0.359
    Sucrose 0.240
    B Products of the invention* 0.001-0.1
    *The product of the invention is obtained e.g. by the extraction process described in Example 13, followed by a drying step.
  • Formulation 22b: Preparation of an Ointment
  • A Excipients
    Low density polyethylene 5.5
    Liquid paraffin qsp 100
    B Products of the invention* 0.001-0.1
    *The product of the invention is obtained e.g. by the extraction process described in Example 13, followed by a drying step.
  • Formulation 22c: Preparation of an Injectable Formula
  • A Excipient
    Isotonic saline solution 5 ml
    B Products of the invention* 0.001-0.1 g
    *The product of the invention is obtained e.g. by the extraction process described in Example 13, followed by a drying step.
  • Example 23 Evaluation of the Cosmetic Acceptance of a Preparation Containing the Subject of the Invention
  • The toxicology tests were performed on the compound obtained according to Example 2, incorporated at a concentration of 10% in a 0.5% xanthan gel, by carrying out an ocular evaluation in the rabbit, by studying the absence of abnormal toxicity by means of a single oral administration in the rat, and by studying the sensitizing power in the guinea-pig.
  • Evaluation of the Primary Cutaneous Irritation in the Rabbit:
  • The preparations described above are applied undiluted in a dose of 0.5 ml to the skin of 3 rabbits according to the method recommended by the OECD directive relating to the study of “the acute irritant/corrosive effect on the skin”.
  • The products are classified according to the criteria defined by the decree of Feb. 1, 1982 published in the OJRF of Feb. 21, 82.
  • The results of these tests afforded the conclusion that the products of the invention were classified as non-irritant on the skin.
  • Evaluation of the Ocular Irritation in the Rabbit:
  • The preparations described above were instilled pure in a single administration of 0.1 ml into the eye of 3 rabbits according to the method recommended by OECD directive no. 405 of 24 Feb. 1987 relating to the study of “the acute irritant/corrosive effect on the eyes”.
  • The results of this test afford the conclusion that the preparations can be considered as non-irritant on the eyes, in terms of directive 91/326 EEC, when used pure or undiluted.
  • Test on the Absence of Abnormal Toxicity by a Single Oral Administration in the Rat:
  • The preparations described were given in a single oral administration in a dose of 5 g/kg body weight to 5 male rats and 5 female rats according to a protocol based on OECD directive no. 401 of 24 Feb. 1987 and adapted to cosmetic products.
  • The LD0 and LD50 are found to be above 5000 mg/kg. The preparations tested are not therefore classified as dangerous preparations on ingestion.
  • Evaluation of the Cutaneous Sensitizing Potential in the Guinea-Pig:
  • The preparations described are subjected to the maximization test described by Magnusson and Kligmann, the protocol being in accordance with OECD directive no. 406.
  • The preparations are classified as non-sensitizing on contact with the skin.

Claims (11)

1. A method for regulating balance between cellular proliferation, differentiation and apoptosis in epithelial cells, comprising applying a pharmaceutical or cosmetic composition onto an epidermis in need thereof, wherein said composition comprises an effective amount of:
i) at least one active ingredient that modulates expression of the lysyl oxidase (LOX) having the amino acid sequence of SEQ ID NO: 1;
ii) at least one active ingredient that modulates expression of the NRAGE having the amino acid sequence of SEQ ID NO: 2; or
iii) a combination of the active ingredient of i) and the active ingredient of ii).
2. The method of claim 1, wherein the epithelial cells are keratinocytes.
3. The method of claim 2, wherein the active ingredient of i) increases the expression of the lysyl oxidase by about 50% in the keratinocytes.
4. The method of claim 2, wherein the active ingredient of i) decreases the expression of the lysyl oxidase by about 50% in the keratinocytes.
5. The method of claim 2, wherein the active ingredient of ii) increases the expression of the NRAGE by about 50% in the keratinocytes.
6. The method of claim 2, wherein the active ingredient of ii) decreases the expression of the NRAGE by about 50% in the keratinocytes.
7. A method of treating aging skin, comprising topically administering onto the skin an effective amount of at least one substance that modulates expression of LOX and/or NRAGE in the skin, said substance is selected from the group consisting of a Sambucus nigra extract, a Humulus lupulus extract, a Ribes rubrum extract, a Cassia amara extract, and an Arbutus unedo extract.
8. A method for treating disorders relating to epidermal hypoproliferation, comprising topically applying an effective amount of a substance that stimulates expression of the LOX having the amino acid sequence of SEQ ID NO: 1 and at least one cosmetically or pharmaceutically acceptable excipient.
9. The method of claim 8, wherein the substance is a Sambucus nigra extract, a Humulus lupulus extract, a Ribes rubrum extract, a Cassia amara extract, or an Arbutus unedo extract.
10. The method of claim 8, further comprising topically applying an effective amount of a substance that inhibits expression of the NRAGE having the amino acid sequence of SEQ ID NO: 2.
11. A cosmetic or pharmaceutical composition for modulating the interaction between LOX and NRAGE proteins in an epidermis, said composition comprising at least one substance selected from the group consisting of a Sambucus nigra extract, a Humulus lupulus extract, a Ribes rubrum extract, a Cassia amara extract, and an Arbutus unedo extract, and at least one cosmetically or pharmaceutically acceptable excipient.
US14/969,783 2005-10-28 2015-12-15 Substance for restoring normal co-expression and interaction between the lox and nrage proteins Abandoned US20160095815A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/969,783 US20160095815A1 (en) 2005-10-28 2015-12-15 Substance for restoring normal co-expression and interaction between the lox and nrage proteins

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
FR0511112A FR2892635B1 (en) 2005-10-28 2005-10-28 SUBSTANCE FOR RESTORING NORMAL CO-EXPRESSION AND INTERACTION BETWEEN LOX AND NRAGE PROTEINS
FRFR0511112 2005-10-28
PCT/FR2006/051117 WO2007048985A2 (en) 2005-10-28 2006-10-26 Substance for restoring normal co-expression and interaction between lox and nrage proteins
US9176808A 2008-11-26 2008-11-26
US13/455,366 US9308161B2 (en) 2005-10-28 2012-04-25 Substance for restoring normal co-expression and interaction between the LOX and NRAGE proteins
US14/969,783 US20160095815A1 (en) 2005-10-28 2015-12-15 Substance for restoring normal co-expression and interaction between the lox and nrage proteins

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US13/455,366 Division US9308161B2 (en) 2005-10-28 2012-04-25 Substance for restoring normal co-expression and interaction between the LOX and NRAGE proteins

Publications (1)

Publication Number Publication Date
US20160095815A1 true US20160095815A1 (en) 2016-04-07

Family

ID=36997700

Family Applications (3)

Application Number Title Priority Date Filing Date
US12/091,768 Abandoned US20110223264A1 (en) 2005-10-28 2006-10-26 Substancefor Restoring Normal Co-expression and Interaction Between the LOX and NRAGE Proteins
US13/455,366 Active 2026-11-10 US9308161B2 (en) 2005-10-28 2012-04-25 Substance for restoring normal co-expression and interaction between the LOX and NRAGE proteins
US14/969,783 Abandoned US20160095815A1 (en) 2005-10-28 2015-12-15 Substance for restoring normal co-expression and interaction between the lox and nrage proteins

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US12/091,768 Abandoned US20110223264A1 (en) 2005-10-28 2006-10-26 Substancefor Restoring Normal Co-expression and Interaction Between the LOX and NRAGE Proteins
US13/455,366 Active 2026-11-10 US9308161B2 (en) 2005-10-28 2012-04-25 Substance for restoring normal co-expression and interaction between the LOX and NRAGE proteins

Country Status (9)

Country Link
US (3) US20110223264A1 (en)
EP (2) EP2345405A1 (en)
JP (2) JP6058249B2 (en)
KR (2) KR101609487B1 (en)
CN (2) CN105963184A (en)
BR (1) BRPI0617938A2 (en)
ES (1) ES2622114T3 (en)
FR (1) FR2892635B1 (en)
WO (1) WO2007048985A2 (en)

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2892635B1 (en) 2005-10-28 2012-11-09 Engelhard Lyon SUBSTANCE FOR RESTORING NORMAL CO-EXPRESSION AND INTERACTION BETWEEN LOX AND NRAGE PROTEINS
FR2970415B1 (en) * 2011-01-14 2013-02-22 Inst Rech Developpement Ird USE OF SIMALIKALACTONE E AS ANTI-CANCER AGENT
FR3009786B1 (en) 2013-08-23 2017-08-11 Basf Beauty Care Solutions France Sas COSMETIC OR DERMATOLOGICAL USE OF A QUASSIA AMARA EXTRACT
FR3011469B1 (en) * 2013-10-08 2017-11-10 Basf Beauty Care Solutions France Sas COSMETIC AND / OR DERMATOLOGICAL USE OF A HAMAMELIS VIRGINIANA EXTRACT
FR3055546B1 (en) * 2016-09-05 2020-01-10 Laboratoires Clarins COSMETIC COMPOSITION COMPRISING A FRUIT EXTRACT OF ARBUTUS UNEDO.
DE102016216866A1 (en) 2016-09-06 2018-03-08 Henkel Ag & Co. Kgaa Oxidative hair dye with optimized lift performance
JP7139287B2 (en) * 2019-06-10 2022-09-20 株式会社ノエビア Solid oily cleansing cosmetic

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040219240A1 (en) * 2001-06-20 2004-11-04 Babish John G. Anti-inflammatory pharmaceutical compositions for reducing inflammation and the treatment or prevention of gastric toxicity

Family Cites Families (64)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE3368377D1 (en) 1982-04-16 1987-01-29 Nestle Sa Lipid composition for oral, enteral or parenteral feeding
JPS61263925A (en) 1985-05-20 1986-11-21 Kanebo Ltd Carcinostatic adjuvant
JPH04271759A (en) * 1991-02-25 1992-09-28 San Ei Chem Ind Ltd Citrus extract-containing gel
FR2699080B1 (en) 1992-12-11 1995-04-07 Lvmh Rech Use of an extract of Simarouba for the attenuation of skin pigment spots or to strengthen the protective function of the skin, or for the preparation of a culture medium for skin cells, and composition thus obtained.
WO1994023732A1 (en) * 1993-04-08 1994-10-27 Kambourakis Medicinal Solutions Pty. Limited Therapeutic preparations containing cypress tree extracts and methods of using same
US5888984A (en) * 1994-05-12 1999-03-30 Dermal Research Laboratories, Inc. Pharmaceutical composition of complex carbohydrates and essential oils and methods of using the same
FR2723096B1 (en) * 1994-07-28 1996-10-18 Univ Picardie ESTERS ASSOCIATING PHENYLACETIC, 3-PHENYL-PROPIONIC, 4-PHENYL-BUTYRIC AND N-BUTYRIC ACIDS WITH D-MANNOSE, XYLITOL AND DERIVATIVES THEREOF. APPLICATIONS AS MEDICINES
EP1059086A1 (en) * 1996-11-05 2000-12-13 Malika H. Haque Use of sandal wood oil or constituents of sandal wood oil for the prevention and treatment of warts, skin blemishes and other viral-induced tumors
JPH11147832A (en) * 1997-11-18 1999-06-02 Noevir Co Ltd Neutrophil esterase inhibitor
IN183330B (en) * 1998-03-23 1999-11-20 Dalmia Ct For Biotechnology
FR2779348B1 (en) * 1998-06-04 2001-06-08 Fabre Pierre Dermo Cosmetique NOVEL PHARMACEUTICAL OR COSMETIC COMPOSITION, IN PARTICULAR HAIR AND PROCESS FOR THE COSMETIC TREATMENT OF CONDITIONS RELATING TO ANGIOGENIC DISORDERS
CN1251302A (en) * 1998-10-19 2000-04-26 郭士元 Traditional Chinese medicine of compounded chlorophyll for treating vegetative nervous disorder and its producing method
DE19853425A1 (en) * 1998-11-19 2000-05-25 Martin Roecken Topical composition containing khellin or its derivatives, useful for phototherapy of psoriasis and eczema, has low mitogenic potential
JP2000169383A (en) * 1998-12-07 2000-06-20 Ichimaru Pharcos Co Ltd Antiallergic agent
JP4271759B2 (en) 1999-01-27 2009-06-03 株式会社岡村製作所 Hanging conveyor
JP2000256345A (en) * 1999-03-02 2000-09-19 T Hasegawa Co Ltd Production of polyphenol compounds
US20010024664A1 (en) * 1999-03-19 2001-09-27 Obukowicz Mark G. Selective COX-2 inhibition from edible plant extracts
GB2370840A (en) * 1999-09-16 2002-07-10 Univ Mcgill Nrage nucleic acids and polypeptides and uses thereof
AU6976300A (en) * 1999-09-16 2001-04-17 Mcgill University Methods for modulating cell proliferation
WO2001026617A1 (en) 1999-10-08 2001-04-19 Coty B.V. Cosmetic preparation of active substances with a synergistically increased radical protection factor
JP2001151634A (en) 1999-11-25 2001-06-05 Lion Corp Composition for external use
JP2001172157A (en) * 1999-12-17 2001-06-26 Shiseido Co Ltd Gelatinase activity inhibitor and anti-aging skin preparation for external use
JP2001253830A (en) 2000-03-13 2001-09-18 Lion Corp Hyaluronidase inhibitor
AU2001261194A1 (en) 2000-05-03 2001-11-12 University Of Hawaii Novel members of the lysyl oxidases family of amine oxidases related applications
FR2810242B1 (en) * 2000-06-16 2003-01-17 Nuxe Lab COSMETIC AND / OR DERMATOLOGICAL COMPOSITION BASED ON COCOA EXTRACTS
JP2002020225A (en) * 2000-07-10 2002-01-23 Naris Cosmetics Co Ltd Humectant composition
US6780442B2 (en) * 2000-08-31 2004-08-24 Hauser, Inc. Efficient method for producing compositions enriched in anthocyanins
KR100651650B1 (en) * 2000-10-12 2006-11-30 한국화학연구원 Anti-cancer composition comprising sesquiterpene compounds isolated from ferulae resina
FR2817747B1 (en) * 2000-12-11 2004-12-03 Oreal USE OF AT LEAST ONE SAPOGENIN, OR A NATURAL EXTRACT CONTAINING IT, TO PREVENT THE SIGNS OF SKIN AGING
KR20030074690A (en) 2000-12-28 2003-09-19 가부시키가이샤 시세이도 Agents for inhibiting or restoring skin damage caused by drying and method of evaluating the same
JP4524935B2 (en) * 2001-02-16 2010-08-18 日油株式会社 Whitening cosmetics
KR20020074746A (en) * 2001-03-21 2002-10-04 주식회사 아미노젠 Anticancer agents containing antigenotoxic and immunostimulation peptides produced from the hydrolyate of silkworm cocoon
FR2824334B1 (en) 2001-05-03 2003-10-10 Coletica METHOD FOR TESTING A SUBSTANCE POSSIBLY ACTIVE IN THE FIELD OF LIPOLYSIS AND ITS MAINLY COSMETIC USE
DE10132953A1 (en) * 2001-07-06 2003-01-16 Beiersdorf Ag Use of hops or Hop-malt extracts in cosmetic or dermatological preparations for the prophylaxis against and treatment of degenerative skin symptoms
JP2003081850A (en) * 2001-09-14 2003-03-19 Maruzen Pharmaceut Co Ltd Skin cosmetic
US20030152588A1 (en) * 2002-01-14 2003-08-14 Hsu-Shan Huang Chinese traditional medicines for psoriasis
JP4393777B2 (en) * 2002-03-19 2010-01-06 株式会社ファンケル Anti-Helicobacter pylori composition
FR2837702B1 (en) * 2002-03-26 2005-01-14 Clarins Lab COSMETIC COMPOSITION FOR SKIN CARE PARTICULARLY AS NIGHT CARE
KR20030079492A (en) * 2002-04-04 2003-10-10 주식회사 바이오시너젠 A Pharmaceutical Composition for Prevention or Treatment of a Disease associated with Inflammation Comprising Erythro-1(4'-methoxyphenyl)-1,2- propanediol or an Extract of Illicium verum Hook FIL containing it
JP2004002237A (en) * 2002-05-31 2004-01-08 Noriko Yagi Anti-aging herb
CN1188118C (en) * 2002-07-23 2005-02-09 中国科学院昆明植物研究所 Medicine for preventing or treating cancer and its usage
GB2393906A (en) * 2002-10-10 2004-04-14 Pharma International Pt Herose Psoriasis treatment
JP2004149729A (en) * 2002-10-31 2004-05-27 Ikeda Shokken Kk Antioxidant
JPWO2004045632A1 (en) * 2002-11-12 2006-03-16 株式会社カネカ Peroxisome proliferator-responsive receptor ligand agent
FR2847267B1 (en) 2002-11-19 2006-07-28 Coletica METHOD FOR TESTING THE ACTIVITY OF A POTENTIALLY ACTIVE SUBSTANCE FOR INHIBITING THE ENZYMATIC ACTIVITY OF A2 PHOSPHOLIPASE
CN100366261C (en) * 2002-12-10 2008-02-06 田中雅也 Skin material for external use and antiprutiric agent for external use and wrinkle-reducing instrument using the same
KR100467081B1 (en) * 2002-12-16 2005-01-24 주식회사 코리아나화장품 Cosmetic Composition for Improving Skin Wrinkles Comprising Bergenia Emeiensis as Active Ingredient
DE10301632A1 (en) * 2003-01-17 2004-07-29 Beiersdorf Ag Cosmetic and dermatological composition, useful e.g. for restructuring and rejuvenating the skin, contains soya bean germ extract, creatinine and creatine
FR2849992B1 (en) * 2003-01-17 2007-03-16 Inst Phytoceutic COMPOSITION FOR ORAL ADMINISTRATION CONTAINING CAPSAICINOIDS
CN1215830C (en) * 2003-03-19 2005-08-24 湖南师范大学 Freckle-diminishing wrinkle-preventing nature white skin-care cream
FR2855969B1 (en) * 2003-06-13 2012-11-30 Coletica STIMULATION OF THE ACTIVITY OF A LYSYL OXIDASE ISOFORM TO FIGHT CERTAIN PATHOLOGIES DUE TO AN INCOMPLETE, ABSENT OR DISORGANIZED ELASTOGENESIS
FR2857588B1 (en) * 2003-07-17 2007-11-30 Oreal USE OF BETA-ENDORPHINE, AGENTS EXERCISING BETA-ENDORPHINE-LIKE ACTIVITY IN COSMETICS AND DERMATOLOGY
CN1589818A (en) * 2003-09-04 2005-03-09 朱桂珍 Internal and external dual purpose Chinese medicine
ES2235642B2 (en) 2003-12-18 2006-03-01 Gat Formulation Gmbh CONTINUOUS MULTI-MICROENCAPSULATION PROCESS FOR THE IMPROVEMENT OF STABILITY AND STORAGE OF BIOLOGICALLY ACTIVE INGREDIENTS.
US7914831B2 (en) * 2004-02-27 2011-03-29 Metaproteomics, Llc Synergistic anti-inflammatory pharmaceutical compositions and related methods using curcuminoids or methylxanthines
EP2279736A1 (en) 2004-03-12 2011-02-02 Life Science Investments Ltd. Microbicidal and antiparasitic compositions comprising quassinoids or quassionid-containing plant extracts
MY142017A (en) * 2004-03-19 2010-08-16 Nestec Sa Composition comprising all essential nutrients of a fruit or a plant material with increased stability and bioavailability and process of forming the same
US20080095866A1 (en) * 2004-09-14 2008-04-24 Ajinomoto Omnichem S.A. Topical Compositions Containing Phosphorylated Polyphenols
CN100518754C (en) * 2005-07-22 2009-07-29 山东省医学科学院药物研究所 Extractive from Gypsophila oldhamiana Miq with anti-cancer activity, prepn. method and application therewith
FR2892635B1 (en) 2005-10-28 2012-11-09 Engelhard Lyon SUBSTANCE FOR RESTORING NORMAL CO-EXPRESSION AND INTERACTION BETWEEN LOX AND NRAGE PROTEINS
WO2008142619A1 (en) * 2007-05-17 2008-11-27 Izun Pharmaceuticals Corporation Use of herbal compositions in the protection and enhancement of extracellular matrix components
WO2009002500A1 (en) 2007-06-25 2008-12-31 Smith, Walter, P. Methods and compositions for reducing the appearance of dynamic facial wrinkles
US20090028895A1 (en) 2007-07-27 2009-01-29 Smith Walter P Methods and compositions for reducing facial lines and wrinkles
FR3009786B1 (en) 2013-08-23 2017-08-11 Basf Beauty Care Solutions France Sas COSMETIC OR DERMATOLOGICAL USE OF A QUASSIA AMARA EXTRACT

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040219240A1 (en) * 2001-06-20 2004-11-04 Babish John G. Anti-inflammatory pharmaceutical compositions for reducing inflammation and the treatment or prevention of gastric toxicity

Also Published As

Publication number Publication date
CN101365421A (en) 2009-02-11
WO2007048985A2 (en) 2007-05-03
EP1948128A2 (en) 2008-07-30
US9308161B2 (en) 2016-04-12
KR101609487B1 (en) 2016-04-05
FR2892635B1 (en) 2012-11-09
KR101500755B1 (en) 2015-03-17
EP1948128B1 (en) 2017-03-15
FR2892635A1 (en) 2007-05-04
KR20080100333A (en) 2008-11-17
US20110223264A1 (en) 2011-09-15
EP2345405A1 (en) 2011-07-20
WO2007048985A3 (en) 2008-04-10
JP2009521401A (en) 2009-06-04
ES2622114T3 (en) 2017-07-05
CN101365421B (en) 2016-07-13
JP2015028036A (en) 2015-02-12
US20120225141A1 (en) 2012-09-06
KR20140083999A (en) 2014-07-04
CN105963184A (en) 2016-09-28
JP6058249B2 (en) 2017-01-11
BRPI0617938A2 (en) 2011-08-09

Similar Documents

Publication Publication Date Title
US20160095815A1 (en) Substance for restoring normal co-expression and interaction between the lox and nrage proteins
US8906425B2 (en) Stimulation of the synthesis of the activity of an isoform of lysyl oxidase-like LOXL for stimulating the formation of elastic fibers
JP6215364B2 (en) MC-1R, MC-2R, and μ opioid receptor modulation
KR100844914B1 (en) Stimulation of the activity of an isoform of lysyl oxidase for combating against some pathologies due to an incomplete, absent or disorganized elastogenesis
JP6059859B2 (en) Stimulation of synthesis and activity of LOXL (lysyl oxidase-like) isoforms to stimulate the formation of elastic fibers
JP4527451B2 (en) Stimulation of synthesis and activity of LOXL (lysyl oxidase-like) isoforms to stimulate the formation of elastic fibers
KR100889460B1 (en) Composition for enhancing melanogenesis in hair containing gdnf, method for enhancing melanogenesis in hair using gdnf, and method for screening hair-melanogenesis enhancer using gdnf expression level
GB2438999A (en) Screening for substances that promote the activity or formation of LOXL
EP1802270B1 (en) Dermatological and/or cosmetic composition containing polypeptides
WO2022210296A1 (en) Oxytocin signal enhancer and oxytocin-induced keratinocyte proliferation-promoting agent
JP4954450B2 (en) Induction of lysyl oxidase isoform activity to address disease states due to failure, loss or disorder of elastic fiber formation
JP2015147798A (en) Stimulation of synthesis and of activity of isoform of loxl (lysyl oxidase-like) for stimulating formation of elastic fiber

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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