WO2015169994A1 - Composition for modulating the activity of sidt1 for the treatment of ifn-mediated infectious, inflammatory or autoimmune diseases - Google Patents

Composition for modulating the activity of sidt1 for the treatment of ifn-mediated infectious, inflammatory or autoimmune diseases Download PDF

Info

Publication number
WO2015169994A1
WO2015169994A1 PCT/ES2015/070369 ES2015070369W WO2015169994A1 WO 2015169994 A1 WO2015169994 A1 WO 2015169994A1 ES 2015070369 W ES2015070369 W ES 2015070369W WO 2015169994 A1 WO2015169994 A1 WO 2015169994A1
Authority
WO
WIPO (PCT)
Prior art keywords
sidt1
ifn
inflammatory
autoimmune
syndrome
Prior art date
Application number
PCT/ES2015/070369
Other languages
Spanish (es)
French (fr)
Inventor
Concepción MARAÑÓN LIZANA
Marta ALARCÓN RIQUELME
Casimiro CASTILLEJO LÓPEZ
Céline COPPARD
Nieves VARELA HERNÁNDEZ
Original Assignee
Fundación Pública Andaluza Progreso Y Salud
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 Fundación Pública Andaluza Progreso Y Salud filed Critical Fundación Pública Andaluza Progreso Y Salud
Publication of WO2015169994A1 publication Critical patent/WO2015169994A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/117Nucleic acids having immunomodulatory properties, e.g. containing CpG-motifs

Definitions

  • Composition capable of modulating the activity of SIDT1 for the treatment of infectious inflammatory or autoimmune diseases mediated by IFN.
  • the present invention is within the field of molecular biology and medicine, and specifically refers to a composition capable of modulating the activity of SIDT1 for the treatment of infectious, inflammatory or interferon-mediated autoimmune diseases (IFN), preferably IFN type I and III, and more specifically for the treatment of psoriasis and lupus. It also refers to a method of selecting drugs useful in the treatment of said diseases and a method for the collection of data useful in the diagnosis of said diseases.
  • IFN infectious, inflammatory or interferon-mediated autoimmune diseases
  • SLE Systemic lupus erythematosus
  • the described genetic associations as well as the study of animal models and biological samples of patients with SLE have allowed the identification of several biological routes involved in the induction of the pathological phenomena characteristic of SLE, such as apoptotic cell clearance, processing of immunocomplexes, the function of Toll type receptors (TLR), or the production of type I interferons (especially IFNa), as well as their response to them.
  • apoptotic cell clearance processing of immunocomplexes
  • TLR Toll type receptors
  • IFNa type I interferons
  • DC Dendritic cells
  • APCs professional antigen presenting cells
  • They are the only APCs capable of stimulating naive T lymphocytes [Villadangos, JA and P. Schnorrer, 2007. Nat Rev Immunol 7 (7): p. 543-555.] And, therefore, have a central role in inducing the autoimmune response.
  • These cells have developed exclusive and probably predominant cross-presentation pathways, which allow them to be restricted to class I molecules of exogenous antigens, after internalization by endocytosis of phagocytosis [Rock, KL, 2003. Nat Immunol 4 (10): p . 941-3].
  • DC can mediate very different effects, such as immunity or tolerance, depending on the subpopulation considered and the microenvironment in which they are located [Steinman, RM, et al., 2003. Annu Rev Immunol, 21: p. 685-711].
  • human DC can be divided into two main subpopulations: conventional myeloid-type (cDC) DCs secrete large amounts of IL-12 and IL-23 after being stimulated with LPS, double-stranded DNA or microbial lipopeptides, thereby inducing a robust Th1 and CD8 effector responses. Phagocytic and presenting cells are prominent, and can be differentiated in vitro from circulating monocytes (MoDC). On the other hand, DC plasmacytoids (pDC) have been classically considered as poor immune system stimulators. However, recent results have shown that peptide-pulsed, or virus-infected, pDCs can effectively stimulate an effector T response [Lui, G., et ai, 2009.
  • Type I interferons possess other important immunoregulatory functions, including stimulation of T lymphocytes [Bracci, L., et al., 2008. Expert Rev Vaccines 7 (3): p. 373-81].
  • the ability of any type of DC to induce immunity or tolerance is a consequence of the displacement of the balance between inhibitory or stimulatory factors, determined by the signals of the microenvironment in which the processing and presentation of the antigen occurs.
  • the final phenotype of DC is reflected by the expression of costimulation molecules and cytokine secretion, for which the balance between IL-23, IL-12 and IL-10 is decisive for the polarization of naive T lymphocytes towards a Th1 / Th2 / Th17 / Treg phenotype.
  • pDC but not cDC
  • pDC can promote differentiation of memory B cells to plasma cells, through an IFNa-dependent mechanism [Poeck , H., et al., 2004. Blood 103 (8): p. 3058-64.]
  • CD70-CD27 interaction Shaw, J., et al., 2010. Blood 115 (15): p. 3051-7].
  • SIDT1 (SID1 transmembrane family, member 1) is a very poorly studied membrane integral protein. Most of the known data on SIDT1 refers to its counterpart in C. elegans, SID1, a membrane protein involved in the mechanism of intercellular iRNA transfer. SID1 is a protein with 11 transmembrane domains, probably involved in the transport of nucleic acids, specifically its overexpression increases the entry of molecules of small RNA interreferencing (siRNA) into human epithelial cells [21-22]. Preliminary results point to a location in the endoplasmic reticulum and in endosomal-lysosomal vesicles of SID1.
  • SIDT1 plays an important role in the recognition of exogenous nucleic acids and in the initiation of the IFN response in pDCs.
  • the authors of the present invention have demonstrated involvement of SIDT1 in the recognition of exogenous nucleic acids and in the initiation of the IFN response in the pDCs, and their inhibition decreases the production of type I and III IFNs.
  • composition of the invention comprising an agent modulating the activity of SIDT1 for the treatment, improvement or prevention of infectious, inflammatory or autoimmune diseases mediated by IFN
  • SIDT1 SID1 transmembrane family, member 1, also called in the literature SID1; SID-1; B830021 E24Rik
  • SIDT1 refers to both the gene and the human SIDT1 protein.
  • SIDT1 is also defined by a nucleotide or polynucleotide sequence, which constitutes the coding sequence of the SIDT1 protein, and which would comprise various variants from: a) nucleic acid molecules encoding a polypeptide comprising the amino acid sequence of SEQ ID NO: 1,
  • nucleic acid molecules whose sequence differs from a) and / or b) due to the degeneracy of the genetic code
  • nucleic acid molecules encoding a polypeptide comprising the amino acid sequence with an identity of at least 80%, 90%, 95%, 98% or 99% with SEQ ID NO: 1. in that the polypeptide encoded by said nucleic acids possesses the activity and structural characteristics of the SIDT1 protein.
  • GenBank GenBank sequence NM_017699.2 or SEQ ID NO: 2.
  • modulating activity refers both to inhibiting (decreasing) or stimulating (increasing) the level of activity of the SIDT1 protein in a cell.
  • the activity of SIDT1 can be modulated by the modification of the levels and / or activity of the SIDT1 protein, or by the modification of the levels to which the SIDT1 gene is transcribed such that the levels of activity of the SIDT1 protein in The cell is modulated.
  • Modulating agents can also be agonists (substances that are capable of binding to a receptor and eliciting a response in the cell, specifically an increase in SIDT1 activity), as antagonists (substances that not only do not activate the receptor, but also actually blocks its activation by agonists). In the context of the present invention, inhibition is the preferred form of modulation.
  • the modulating agents comprised in the composition of the invention are selected from a list comprising: a) an organic molecule, b) an RNA molecule, c) an antisense oligonucleotide, d) an antibody, or e) a ribozyme F) A nuclease capable of editing chromosomal DNA (ZFN, Talen, CRISPR).
  • organic molecules that can specifically bind SIDT1 without binding to other polypeptides or proteins will preferably have a weight of 100 to 20,000 daltons, more preferably 500 to 15,000 daltons, and more preferably 1000 to 10,000 daltons. Bookstores of organic molecules are commercially available.
  • the route of administration may be, without limitation, intraperitoneal, intrathecal, intravenous, intramuscular, subcutaneous, intraventricular, oral, enteral, parenteral, intranasal or dermal.
  • nucleotide sequences specifically complementary to a particular DNA or RNA sequence could form complexes and block transcription or translation.
  • interfering RNA interfering RNA
  • tools have been developed that allow specific inhibition of gene expression.
  • the inhibition of the expression of the SIDT1 protein would therefore constitute the inhibition of its biological activity, and in particular, of the activity that is contributing to the aggravation of the inflammatory or autoimmune characteristics mediated by IFN.
  • antisense polynucleotides are meant ribonucleotide or deoxyribonucleotide chains that can inhibit the production of the SIDT1 protein by one of these three mechanisms:
  • SIDT1 Interfering transcription by hybridizing in the structural gene or in a regulatory region of the gene encoding SIDT1. Since transcription or expression is effectively blocked by hybridization of the antisense oligonucleotide with DNA, the production of SIDT1 decreases. 2- The binding of the antisense oligonucleotide in the cytoplasm with the mRNA, interfering with the formation of the translation construct itself, inhibiting the translation of mRNA into the protein.
  • Antisense oligonucleotides capable of modulating the activity of SIDT1 can be easily synthesized from the known sequence SEQ ID NO: 2 by applying knowledge of the state of the art. For example, and without limiting our, it could be a sequence of ribonucleotides or RNA that belongs to the so-called siRNA (small interfering RNA), small interfering RNA or silencing RNA, capable of inhibiting the genetic expression of the SIDT1 protein.
  • siRNA small interfering RNA
  • small interfering RNA small interfering RNA
  • silencing RNA small interfering RNA
  • siRNA small interfering RNA or small interfering RNA
  • SIDT1 small interfering RNA or small interfering RNA
  • siRNA is understood to be a class of double stranded RNA 19 to 25 nucleotides long, and more preferably between 21 and 23 nucleotides, which is involved in the route of RNA interference, where siRNA interferes with the expression of a specific gene.
  • this specific gene is SIDT1. It could also be any siRNA capable of hybridizing a nucleic acid molecule encoding the human SIDT1 protein that is collected in SEQ ID NO: 1.
  • RNA constructs that contains at least any one of the possible nucleotide sequences of siRNA capable of inhibiting the expression of SIDT1, and without prejudice to the addition of any of the RNA sequences and constructions of the invention described above that are subject to modifications, preferably chemical, leading to greater stability against the action of ribonucleases. and with it to greater efficiency. Without these modifications involving the alteration of its mechanism of action, which is the specific binding to the RISC complex (RNA-induced silencing complex), activating it and manifesting a helicase activity that separates the two strands leaving only the antisense strand associated with the complex.
  • RISC complex RNA-induced silencing complex
  • the resulting ribonucleoproteic complex binds to the target mRNA (messenger RNA of SIDT1, which is collected in SEQ ID NO: 2). If the complementarity is not perfect, RISC is associated with the messenger and the translation is attenuated. But if it is perfect, RISC acts as RNasa, cutting the messenger and being free to repeat the process. Additionally, it is apparent to one skilled in the art that a large number of mRNA polynucleotides can be translated into SIDT1 as a consequence, for example, that the genetic code is degenerated. Any siRNA capable of inhibiting the translation of these mRNAs is also part of the invention.
  • siRNA sequence of the invention or of the RNA construct of the invention would be apparent to one skilled in the art, and could be carried out by chemical synthesis, which also allows the incorporation of chemical modifications in both different nucleotides of the product such as the incorporation of other chemical compounds at any of the ends.
  • the synthesis could also be carried out enzymatically using any of the available RNA polymerases. Enzymatic synthesis also allows some chemical modification of inhibitor products or RNAs.
  • the nucleotide sequence design of the siRNA of the invention would also be apparent to one skilled in the art. Thus, it could be done through a random design in which 19-25 bases of the target mRNA are selected without taking into account the sequence or positional information it has in the transcript.
  • Another non-limiting alternative of the present invention would be the conventional design by simple parameters developed by the pioneers of the art (Calipel, A. et al., 2003. J Biol Chem. 278 (43): 42409-42418) completed with an analysis BLAST nucleotide.
  • Another possibility could be a rational design, in which a computer procedure is used to identify the optimal siRNA targets in an mRNA.
  • a genetic DNA construct could also be part of the composition of the invention, which would direct the in vitro or intracellular transcription of the siRNA sequence or RNA construct of the invention, and comprising at least one of the following types of sequences: a) DNA nucleotide sequence, preferably double stranded, comprising at least the sequence encoding the siRNA of the invention or the RNA construct of the invention for transcription, or, b) nucleotide sequence of DNA, preferably double stranded, corresponding to a gene expression system or vector comprising the sequence coding for the RNA sequence of the invention operably linked with at least one promoter that directs the transcription of said nucleotide sequence of interest, and with other sequences necessary or appropriate for transcription and its appropriate regulation in time and place, for example, start signals and termination, cutting sites, polyadenylation signal, origin of
  • SIDT1 is contributing to the worsening of inflammatory and / or autoimmune characteristics and to the onset of the disease.
  • Multiple of these constructs, systems or expression vectors can be obtained by conventional methods known to those skilled in the art (Sambrook et al. 2001. Molecular Cloning: A laboratory Manual. Coid Spring Harbor laboratory Press, New York)
  • compositions of the present invention allow the transfection of the siRNA of the invention into a cell, in vivo or in vitro. Transfection could be carried out, but not limited to, direct transfection or vectors that facilitate the access of siRNA into the cell.
  • vectors are, without limitation, retroviruses, lentiviruses, adenoviruses, adeno-associated viruses, Herpes simplex viruses, non-viral DNA plasmids, cationic liposomes and molecular conjugates.
  • the siRNAs of the present invention, as well as RNA or DNA precursors of these siRNAs can be conjugated with release peptides or other compounds to favor the transport of these siRNAs into the cell.
  • Nucleases designed to block or activate gene expression are also part of the invention.
  • antibody refers to immunoglobulin molecules and immunologically active portions of immunoglobulin molecules, that is, molecules that contain an antigen binding site that specifically binds (immunoreacts with) the protein. SIDT1.
  • portions of immunologically active immunoglobulin molecules include F (ab) and F (ab ') 2 fragments that can be generated by treating the antibody with an enzyme such as pepsin. It can be a monoclonal or polyclonal antibody.
  • Antibodies capable of binding to the SIDT1 protein can be used to inhibit the activity of said protein. Many such antibodies are commercially available. The antibodies, or fragments thereof, may be able to inhibit the activity of the SIDT1 protein that contributes to the acquisition of the characteristics of inflammatory and / or autoimmune diseases.
  • the antibodies can be polyclonal (typically include different antibodies directed against different determinants or epitopes) or monoclonal (directed against a single determinant in the antigen).
  • the monoclonal antibody can be biochemically altered, by genetic manipulation, or it can be synthetic, possibly lacking the antibody in whole or in parts, of portions that are not necessary for the recognition of SIDT1 and being replaced by others that communicate to the antibody additional advantageous properties.
  • the antibody can also be recombinant, chimeric, humanized, synthetic or a combination of any of the foregoing.
  • a "recombinant antibody or polypeptide” is one that has been produced in a host cell that has been transformed or transfected with the nucleic acid encoding the polypeptide, or produces the polypeptide as a result of homologous recombination.
  • rAc can be expressed and directed to specific cellular sub-compartments when the appropriate sequences for intracellular traffic are incorporated.
  • These antibodies are called intrabodies, and have proven effective not only to divert proteins from their usual compartment or block interactions between proteins involved in signaling pathways, but also to activate intracellular proteins.
  • DNA constructs capable of transcribing to a peptide, antibody or antibody fragment, for use in the treatment, improvement or prevention of infectious, inflammatory or autoimmune diseases mediated by IFN.
  • Said genetic construction of DNA would direct the in vitro or intracellular transcription of the antibody sequence or fragment thereof, and comprises at least one of the following types of sequences: a) DNA nucleotide sequence, preferably double stranded, which It comprises, at least, the coding sequence of the antibody of the invention or of the antibody fragment of the invention for in vitro, or intracellular, transcription, b) DNA nucleotide sequence, preferably double stranded, corresponding to a system or vector of gene expression comprising the sequence coding for the antibody sequence or antibody fragment of the invention operably linked with at least one promoter that directs the transcription of said nucleotide sequence of interest, and with other sequences necessary or appropriate for transcription and its proper regulation in time and place, for example, start and end signals, cut sites, polyadenylation signal, origin of replication
  • ribozyme refers to a catalytic polynucleotide (typically RNA), which can be constructed to specifically recognize, by hybridization, an mRNA and fragment it or eliminate its expression. Ribozymes can be introduced into the cell as catalytic RNA molecules or as genetic constructs that are expressed to RNA catalytic molecules.
  • autoimmune disease refers to a condition in a subject characterized by a cellular, tissue and / or organ injury caused by an immunological reaction of the subject to its own cells, tissues and / or organs.
  • autoimmune diseases include alopecia areata, ankylosing spondylitis, antiphospholipid syndrome, Addison autoimmune disease, autoimmune diseases of the adrenal gland, autoimmune hemolytic anemia, autoimmune hepatitis, autoimmune ooforitis and orchitis, autoimmune thrombocytopenia, autoimmune disease Behcet, bullous pemphigoid, cardiomyopathy, celiac disease, dermatitis, chronic fatigue, immune dysfunction syndrome (CFS), chronic inflammatory demyelinating polyneuropathy, Churg - Strauss syndrome, scarring penfigoid, CREST syndrome, cryogglutinin syndrome, discoid lupus , essential mixed cryoglobulinemia, fibromyalgia-fibromiositis, glomerulonephritis, Graves disease, Guillain-Barre syndrome, Hashimoto's thyroiditis, idiopathic pulmonary fibrosis,
  • treatment means the administration of a compound or formulation according to the invention to prevent, ameliorate or eliminate the disease or one or more symptoms associated with said disease.
  • Treatment also covers the prevention, improvement or elimination of the physiological sequel of the disease.
  • immunosuppressants are substances that decrease the immune response. They can be either exogenous, such as immunosuppressive drugs, or endogenous.
  • immunosuppressive drugs are glucocorticoids, which decreases both the expansion of B cell clones and the synthesis of antibodies; cytostatics, which affects the proliferation of both T cells and B cells, examples of alkylating agents are cytostatic agents, antimetabolites (for example, folic acid analogs, purine analogs, pyrimidine analogs, protein synthesis inhibitors ); antibodies or drugs that act on immunophilins such as cyclosporine, tacrolimus or sirolimus. SLE is treated with immunosuppression, mainly with cyclophosphamide or corticosteroids.
  • the composition of the invention is used for the treatment of an autoimmune disease.
  • the autoimmune disease is selected from the list consisting of: alopecia areata, ankylosing spondylitis, antiphospholipid syndrome, Addison autoimmune disease, autoimmune diseases of the adrenal gland, autoimmune hemolytic anemia , autoimmune hepatitis, autoimmune oophoritis and orchitis, autoimmune thrombocytopenia, Behcet's disease, bullous pemphigoid, cardiomyopathy, celiac disease, dermatitis, chronic fatigue, immune dysfunction syndrome (CFS), chronic inflammatory demyelinating polyneuropathy, Churg-Strauss syndrome , scarring pemphigoid, CREST syndrome, cryogglutinin disease, discoid lupus, essential mixed cryoglobulinemia, fibromyalgia-fibromiositis, glomerulonephritis
  • the autoimmune disease is systemic lupus erythematosus. In another particular embodiment of the invention, the autoimmune disease is psoriasis.
  • composition provided by this invention may be provided by any route of administration, for which said composition will be formulated in the pharmaceutical form appropriate to the route of administration chosen.
  • SIDT1 activity modulating agents of said compositions are in a therapeutically effective amount.
  • therapeutically effective amount refers to the amount of modulating agents (or genetic constructs that allow their intracellular expression) calculated to produce the desired effect and, in general, will be determined, among other causes. , due to the characteristics of said agents (and constructions) and the therapeutic effect to be achieved.
  • Pharmaceutically acceptable adjuvants and vehicles that can be used in said compositions are the vehicles known to those skilled in the art.
  • the invention provides methods for identifying compounds that can be used for the treatment of diseases related to SIDT1, and in particular inflammatory diseases or autoimmune diseases mediated by IFN, and preferably mediated by IFN type I and III.
  • SIDT1 Assays to identify these molecules, compounds or agents that modulate the activity of SIDT1 may employ cells expressing SIDT1, or in assays with isolated SIDT1 (or with its variants, such as biologically active fragments or fusion proteins that include a portion or part of YES DT1).
  • another aspect of the invention consists in a method of selecting therapeutic agents useful in the treatment of inflammatory or autoimmune diseases mediated by IFN, and more specifically for the treatment of psoriasis and lupus; which comprises: a) contacting the compound to be analyzed with the SIDT1 polypeptide, and b) detecting the binding of said compound to be analyzed with the SIDT1 polypeptide.
  • SIDT1 polypeptide Compounds that bind to the SIDT1 polypeptide would be identified as potential therapeutic agents against inflammatory or autoimmune diseases mediated by IFN, and more specifically for the treatment of psoriasis and lupus.
  • these assays may involve the complete SIDT1 polypeptide, a biologically active fragment thereof, or a fusion protein that involves all or a portion of the SIDT1 polypeptide. Determining the ability of a compound to modulate the activity of SIDT1 can be performed, for example, by determining the ability of SIDT1 to bind or interact with a target molecule of said compound, directly or indirectly. They can also be activity tests, directly or indirectly measuring the activity of SIDT1. It can also be an expression assay, directly or indirectly determining the expression of the SIDT1 mRNA or SIDT1 protein.
  • tests can also be combined with an in vivo test by measuring the effect of a test compound on the symptoms of SIDT1-related diseases, and in particular those inflammatory or autoimmune diseases mediated by IFN, and preferably mediated by IFN type I and III (by example, but not limited, on animal models or other model systems known in the art).
  • the compounds to be tested used in the method of selection of therapeutic agents are not limited to low molecular weight organic molecules, proteins (including antibodies), peptides, oliogonucleotides, etc. They can be natural and / or synthetic.
  • antibodies capable of binding to an epitope of SIDT1 which can be used therapeutically, as discussed above, can also be used in immunohistochemical assays, such as Western blots, ELISAs, radioimmunoassays, immunoprecipitation assays, or other immunohistochemical assays known in The state of the art.
  • SIDT1 can be used to immunize an animal, to obtain polyclonal antibodies.
  • Monoclonal antibodies can also be prepared by techniques that allow the production of antibodies by cultured cell lines, including, but not limited to, hybridomas, human B-cell hybridomas or collections of nanobodies. Techniques for producing chimeric, humanized or synthetic antibodies are known.
  • the therapeutic agents identified by the selection method described herein can be used in an animal or other model to determine the mechanism of action of said agent.
  • the therapeutic agents selected by the method described herein would be used in the treatment of diseases that occur with the alteration of SIDT1 and, in particular, IFN-mediated inflammatory or autoimmune diseases, preferably by type I and III IFNs, and more specifically psoriasis and lupus.
  • a method of selection of therapeutic agents useful in the treatment of inflammatory diseases or autoimmune diseases mediated by IFN, preferably by type I and III IFN, and more specifically for the treatment of psoriasis and lupus comprises: a) determining the activity of SIDT1 at an established concentration of the compound to be analyzed or in the absence of said compound, b) determining the activity of SIDT1 at a concentration of the compound to be analyzed different from that of a).
  • Compounds that give rise to a different activity of SIDT1 would be identified as potential therapeutic agents against inflammatory or autoimmune diseases mediated by IFN, preferably by type I and III IFN, and more specifically for psoriasis and lupus.
  • Another aspect of the invention consists in a method of selecting therapeutic agents useful in the treatment of inflammatory or autoimmune diseases mediated by IFN, preferably by type I and III IFN, and more specifically psoriasis and lupus, comprising: a ) contacting the compound to be analyzed with the SIDT1 polynucleotide, b) detecting the binding of said compound to be analyzed with the SIDT1 polynucleotide
  • SIDT1 polynucleotide Compounds that bind to the SIDT1 polynucleotide would be identified as potential therapeutic agents against inflammatory or autoimmune diseases mediated by IFN, preferably by type I and III IFN, and more specifically from psoriasis and lupus.
  • SIDT1 diseases in which the alteration of the activity of SIDT1 can be diagnostic, and in particular inflammatory diseases or autoimmune mediated by IFN, and more specifically psoriasis and lupus, can be detected by measuring the amount of nucleic acids (DNA and / or RNA and / or mRNA) encoding SIDT1, or the amount of SIDT1 protein that is expressed, compared to normal cells.
  • Oligonucleotide detection it can be done by methods well known in the state of the art (such as, but not limited to, probes with labeled nucleotides, DNA-DNA or DNA-RNA hybridization, PCR amplification using labeled nucleotides, RT-PCR).
  • SIDT1 protein Methods for detecting the expression of the SIDT1 protein are also well known in the state of the art, such as, but not limited to, poly or monoclonal antibodies, ELISA, radioimmunoassay (RIA), and FACS (fluorescence activated cell sorting).
  • a method for the collection of data useful in the diagnosis and / or prognosis of inflammatory or autoimmune diseases mediated by IFN, and more specifically psoriasis and lupus comprising: a) determine the expression of SIDT1 in a sample taken from a mammal, b) compare the values of the expression of SIDT1 obtained in a) with the standard values in healthy or diseased mammals.
  • IFN-mediated inflammatory or autoimmune diseases include, but are not limited to, systemic lupus erythematosus, systemic sclerosis, psoriasis, Sjógren syndrome, rheumatoid arthritis, Accardi-Goutiéres syndrome, alopecia areata, ankylosing spondylitis, antiphospholipid syndrome, antiphospholipid syndrome Addison autoimmune disease, autoimmune diseases of the adrenal gland, autoimmune hemolytic anemia, autoimmune hepatitis, autoimmune oophoritis and orchitis, autoimmune thrombocytopenia, Behcet's disease, bullous pemphigoid, cardiomyopathy, celiac disease, dermatitis, chronic fatigue, dysfunction syndrome Immune (CFS), chronic inflammatory demyelinating polyneuropathy, Churg-Strauss syndrome, scarring penfigoid, CREST syndrome, cryoagglutinin disease, discoid lupus, essential mixed cryoglobul
  • peptide refers to a polymeric form of amino acids of any length, which may be coding or non-coding, chemically or biochemically modified.
  • Fig. 1 Expression of the SIDT1 gene in several cell lines.
  • Fig. 2 Negatively regulated genes in Gen2.2 cells silenced for SIDT1, after stimulation with ODN2006 for 6h.
  • the expression of the different genes was carried out by means of expression arrays.
  • the table shows the relative expression in the silenced cells versus the cells treated with a control shRNA, without target.
  • the table shows the first 50 genes sorted by importance of effect.
  • Fig. 3 Production of pro-inflammatory cytokines in Gen2.2 shSIDTI cells after stimulation with TLR ligands) (ODN2006) and TLR7 (Imiquimod). The cells were stimulated for 24h with ODN2006 or imiquimod and the amount of TNFa, IL-6 and IL-8 in the supernatant was measured by ELISA. As a control, non-transduced cells and transduced cells with a control shRNA were included.
  • the gen2.2 shSIDTI cells and their controls were stimulated as in Figure 3 and the production of interferons of type I (IFNal) and III (IFNI1) and of the IFN-dependent chemokine CCL3 was measured by ELISA.
  • Fig. 5 Partial inhibition of oligonucleotide entry into the cell by silencing SIDT1.
  • the gen2.2 shSIDTI cells were incubated with the TLR9 stimulating oligonucleotide labeled ODN2006-Cy5 and their entry into the cell was measured by flow cytometry.
  • non-transduced cells and transduced cells with a control shRNA were included.
  • SIDT1 silencing blocks the production of type I IFN by pDC isolated ex vivo in response to ODN2006.
  • Fig. 7 Effect of SIDT1 silencing on type I IFN secretion after stimulation of DNA and RNA endosomal and cytosolic receptors.
  • Gen2.2 shSIDTI cells and their controls were stimulated with ODN2006 or poly l: C in the presence or absence of lipofectamine.
  • Type I IFN activity was measured in the supernatant after 24h of stimulation by the use of indicator cells.
  • Fig. 8 Phosphorylation of TBK-1 and F2a after stimulation of TLR9.
  • Gen2.2 shSIDTI cells and their controls were stimulated with ODN2006 and phosphorylation of TBK-1 and elF2a was measured by Western blot. Actin protein was used to estimate the amount of total proteins in each of the lanes.
  • Fig. 1A lymphoid-type hematopoietic cell lines
  • Fig. 1A T and B lymphocytes, NK cells and plasmacytoid dendritic cells
  • pDCs plasmacytoid dendritic cells
  • His study focuses on pDCs, which are key cells for the organization of the immune response, because they are antigen presenting cells with a high capacity for cytokine production in response to microbial stimuli.
  • the pDCs detect infectious agents or dead cells through exogenous nucleic acid endosomal receptors, which recognize RNA sequences (Toll-like receptor 7, TLR7), or DNA with non-methylated CpG motifs (TLR9).
  • the pDCs are known as the natural interferon-producing cells (IFN), due to their very high capacity to produce large amounts of type I (a and ⁇ ) and III ( ⁇ ) IFN, fundamental cytokines for controlling infection, as well as for Modulate the activity of T and B lymphocytes.
  • IFN interferon-producing cells
  • a and ⁇ type I
  • III
  • Gen2.2 cell line obtained from a patient with a Neoplasm with pDC phenotype.
  • This cell line is currently the main cell model for the study of the biology of the pDC.
  • the SIDT1 mRNA is induced after stimulation with TLR7 (imiquimod) and TLR9 (ODN2006) ligands (Figure 1B) suggesting a role of SIDT1 in the response mediated by these receptors.
  • Expression arrays were performed using Gen2.2 cells transduced with lentiviral vectors carrying shRNA sequences specific for SIDT1, using shRNA control without target.
  • the repressed genes in shSIDTI cells after stimulation with ODN2006 are enriched in genes related to the immune system, specifically with the IFN pathway.
  • SIDT1 is preferentially expressed in lymphoid line cell lines: T and B lymphocytes, NK cells and plasmacytoid dendritic cells (pDC). Given the central role of regulating the immune system of the pDCs, this cell type was chosen as a model to study the function of SIDT1.
  • Gen2.2 cells were stimulated with ligands of the endosomal sensors of TLR-7 (imiquimod 3 ⁇ g / ml) and TLR-9 nucleic acids (ODN2006, 1 mM) for 24 and 48h.
  • the results of qPCR in Figure 1 B show a strong overexpression of SIDT1 after stimulation, indicating a role of SIDT1 in the immune response mediated by these receptors.
  • transductions were performed with lentiviral vectors carrying three shRNA sequences specific for SIDT1 (shRNA # 1 AGCCGTGAGGGTGTATGTGAA, (SEQ ID NO: 3 ); shRNA # 2 CTACTTGGGATGCCTTCTTGT (SEQ ID NO: 4); shRNA # 3 GTGCATTCTGCTGGATTTCTT (SEQ ID NO: 5)), cloned into the vector pLenti-H1 (AMSBIO).
  • a lentivirus was used carrying a shRNA sequence without a target (shCTL GTCTCCACGCGCAGTACATTT (SEQ ID NO: 6)).
  • a multiplicity of infection (moi) of 1 was used.
  • Gen2.2 cells and cells transduced with The three shRNAs specific for SIDT1 (shSIDTI) as their control shRNA (shCTL) were stimulated for 6h with 1 ⁇ ODN2006.
  • RNA was purified after stimulation and was used for studies of RNA expression arrays.
  • the raw data were generated using the GenomeStudio software (lllumina) and subsequently analyzed in the R and Bioconductor statistical environment.
  • the expression values of each probe were determined after correction of the background signal using the negative controls and subsequently quantile normalization was applied to eliminate technical variations.
  • Probes with detection values p ⁇ 0.01 in at least 10% of the samples were removed from the analysis and the average value of the expression of each gene was computed by applying the median to the value of all probes encoding the same gene. Differential expression analysis was carried out using the Limma package. Genes that showed a corrected p-value less than 0.05 were considered statistically significant.
  • Figure 50 shows the 50 most repressed genes in Gen2.2 shSIDTI cells stimulated in relation to shCTL.
  • Circulating pDCs were isolated from blood units from healthy donors using the BDCA-4 Positive Selection Kit in an Automacs Pro separator (both from Miltenyi Biotec), following the manufacturer's instructions. In all cases, pDCs with more than 90% purity were obtained.
  • the purified pDCs were transduced with the shSIDTI or shCTL lentiviruses at a moi of 0.4 and were stimulated with 1 M ODN2006 in RPMI medium supplemented with 5% autologous inactivated plasma for 24 h. SIDT1 expression was quantified using the Quantigene Assay Kit (Panomics / Affimetrix) and the Glomax Multidetector System (Promega) luminometer, following the manufacturers instructions.
  • SIDT1 is limited to endosomal nucleic acid receptors, or on the contrary is also involved in the IFN response mediated by cytosolic receptors
  • stimulations with ODN2006 and poly l: C encapsulated in liposomes were performed through use. of lipofectamine, to allow the access of the ligands to the cytosol.
  • Gen2.2 cells silenced for SIDT1 also showed an altered response after stimulation of cytosolic DNA receptors with CpG + lipofectamine.
  • cytosolic RNA receptors poly 1: C + lipofectamine
  • shSIDTI cells which SIDT1 is not a general mediator of the IFN response to exogenous nucleic acids, but is a selective mediator of certain recognition pathways in the pDC.
  • TBK-1 activates the phosphorylation of different members of the IRF family, allowing its translocation to the nucleus to activate IFN signature genes, and therefore It is essential for the development of a complete response mediated by TLR9.
  • the translation initiation factor of mRNA elF2a is involved in the response to stimulation of TLR9 and TLR3.
  • stimulation with ODN2006 induces phosphorylation of TBK-1 and elF2a.
  • SIDT1 is essential for the establishment of an optimal signal transduction cascade for the induction of type I and III interferons after stimulation of TLR7, TLR9 and cytosolic DNA receptors.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Molecular Biology (AREA)
  • Biotechnology (AREA)
  • Chemical & Material Sciences (AREA)
  • Wood Science & Technology (AREA)
  • Organic Chemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Zoology (AREA)
  • General Health & Medical Sciences (AREA)
  • Physics & Mathematics (AREA)
  • Microbiology (AREA)
  • Plant Pathology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Immunology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention relates to a composition for modulating the activity of SIDT1 for the treatment of infectious, inflammatory or autoimmune diseases mediated by interferons (IFN), preferably type I and type III IFN, more specifically for the treatment of psoriasis and lupus. The invention also relates to a method for selecting drugs that can be used in the treatment of said diseases and to a method for collecting data that can be used in the diagnosis of said diseases.

Description

Composición capaz de modular la actividad de SIDT1 para el tratamiento de enfermedades infecciosas inflamatorias o autoinmunes mediadas por IFN.  Composition capable of modulating the activity of SIDT1 for the treatment of infectious inflammatory or autoimmune diseases mediated by IFN.
Campo técnico Technical field
La presente invención se encuentra dentro del campo de la biología molecular y de la medicina, y específicamente se refiere a una composición capaz de modular la actividad de la SIDT1 para el tratamiento de enfermedades infecciosas, inflamatorias o autoinmunes mediadas por interferones (IFN), preferiblemente IFN de tipo I y III, y más específicamente para el tratamiento de la psoriasis y el lupus. También se refiere a un método de selección de fármacos útiles en el tratamiento de dichas enfermedades y a un método para la recolección de datos útiles en el diagnóstico de dichas enfermedades. The present invention is within the field of molecular biology and medicine, and specifically refers to a composition capable of modulating the activity of SIDT1 for the treatment of infectious, inflammatory or interferon-mediated autoimmune diseases (IFN), preferably IFN type I and III, and more specifically for the treatment of psoriasis and lupus. It also refers to a method of selecting drugs useful in the treatment of said diseases and a method for the collection of data useful in the diagnosis of said diseases.
Antecedentes de la invención Background of the invention
El lupus eritematoso sistémico (SLE) es una enfermedad crónica autoinmune, caracterizada por la producción de autoanticuerpos y la inducción de daño tisular mediada por la deposición de inmunocomplejos autoanticuerpo-autoantígeno. Esta enfermedad deriva en complicaciones serias de tipo renal, cardiovascular y neurológico. La patogénesis del SLE implica interacciones complejas de múltiples genes y factores medioambientales. Hasta la fecha se han descrito varios genes implicados en la susceptibilidad a SLE (Harley, IT and Kaufman, C.D. 2009. Nat Rev 10(5): p. 285-90), sin embargo el entendimiento de cómo las variantes génicas identificadas afectan el riesgo y la progresión del SLE es todavía insuficiente. Las asociaciones genéticas descritas, así como el estudio de modelos animales y de muestras biológicas de pacientes con SLE han permitido la identificación de varias rutas biológicas implicadas en la inducción de los fenómenos patológicos característicos del SLE, como son el aclaramiento de células apoptóticas, el procesamiento de inmunocomplejos, la función de los receptores tipo Toll (TLR), o la producción de interferones de tipo I (sobre todo IFNa), así como su respuesta a los mismos. Systemic lupus erythematosus (SLE) is a chronic autoimmune disease, characterized by the production of autoantibodies and the induction of tissue damage mediated by the deposition of autoantibody-autoantigen immune complexes. This disease results in serious complications of renal, cardiovascular and neurological type. The pathogenesis of SLE involves complex interactions of multiple genes and environmental factors. To date, several genes involved in susceptibility to SLE have been described (Harley, IT and Kaufman, CD 2009. Nat Rev 10 (5): p. 285-90), however the understanding of how the identified gene variants affect the Risk and progression of SLE is still insufficient. The described genetic associations, as well as the study of animal models and biological samples of patients with SLE have allowed the identification of several biological routes involved in the induction of the pathological phenomena characteristic of SLE, such as apoptotic cell clearance, processing of immunocomplexes, the function of Toll type receptors (TLR), or the production of type I interferons (especially IFNa), as well as their response to them.
Las células dendríticas (DC) son células presentadoras del antígeno (APC) profesionales, situadas en la interfase entre la respuesta inmune innata y adaptativa. Son las únicas APC con capacidad para estimular los linfocitos T naive [Villadangos, J.A. and P. Schnorrer, 2007. Nat Rev Immunol 7(7): p. 543-555.] y, por lo tanto, tienen un papel central en la inducción de la respuesta autoinmune. Estas células han desarrollado vías exclusivas y probablemente predominantes de presentación cruzada, que les permiten la presentación restringida a moléculas de clase I de antígenos exógenos, tras su internalización mediante endocitosis of fagocitosis [Rock, K.L., 2003. Nat Immunol 4(10): p. 941-3]. Estas vías son cruciales para la estimulación de una respuesta específica de antígenos que no son producidos en el citosol de las DC, como son aquellos reconocidos en el caso de las enfermedades autoinmunes [Segura, E. and J.A. 2009. Curr Opin Immunol 21 (1): p. 105-10]. Las DC pueden mediar efectos muy diferentes, como inmunidad o tolerancia, dependiendo de la subpoblación que se considere y el microambiente en el que se encuentren [Steinman, R.M., et al., 2003. Annu Rev Immunol, 21 : p. 685-711]. las DC humanas pueden dividirse en dos subpoblaciones principales: las DC convencionales (cDC), de tipo mieloide, secretan grandes cantidades de IL-12 e IL-23 tras ser estimuladas con LPS, DNA de doble cadena o lipopéptidos microbianos, induciendo por tanto un respuestas de tipo Th1 y CD8 efectoras robustas. Son destacadas células fagocíticas y presentadoras, y pueden diferenciarse in vitro a partir de monocitos circulantes (MoDC). Por otro lado, las DC plasmacitoides (pDC) se han considerado clásicamente como pobres estimuladoras del sistema inmune. Sin embargo, los resultados recientes han demostrado que las pDC pulsadas con péptidos, o infectadas con virus, pueden estimular eficazmente una respuesta T efectora [Lui, G., et ai, 2009. PLoS One 4(9): p. e71 11 ; Di Pucchio, T., et al., 2008. Nat Immunol 9(5): p. 551-7.], pero también activar los linfocitos T reguladores CD4+ o CD8+ [Moseman, E.A., et al., 2004. J Immunol 173(7): p. 4433-42]. Aunque se les suponía incapaces de realizar la presentación cruzada, se ha demostrado que las pDC circulantes humanas realizan ex vivo la presentación cruzada de antígenos de HIV tras la fagocitosis de células infectadas [Hoeffel, G., et al., 2007. Immunity 27(3): p. 481-92]. Producen grandes cantidades de IFN de tipo I y III en respuesta a virus, fragmentos de DNA ricos en motivos CpG no metilados, o ligandos de TLR-7. Las pDC estimulan la diferenciación de las células plasmáticas a través de IFN de tipo I e IL-6 [Jego, G., et al., 2003. Immunity 19(2): p. 225-34]. Los interferones de tipo I poseen otras importantes funciones inmunorreguladoras, incluyendo la estimulación de los linfocitos T [Bracci, L., et al., 2008. Expert Rev Vaccines 7(3): p. 373-81]. La capacidad de cualquier tipo de DC para inducir inmunidad o tolerancia es consecuencia del desplazamiento del equilibrio entre factores inhibidores o estimuladores, determinados por las señales del microambiente en el que ocurre el procesamiento y presentación del antígeno. El fenotipo final de las DC se ve reflejado por la expresión de moléculas de coestimulación y la secreción de citoquinas, para las que el equilibrio entre IL-23, IL-12 e IL-10 es determinante para la polarización de los linfocitos T naive hacia un fenotipo Th1/Th2/Th17/Treg. Aunque la interacción entre las DC y los linfocitos B ha sido menos estudiada, se ha observado que las pDC, pero no las cDC, pueden promover la diferenciación de las células B memoria a células plasmáticas, a través de un mecanismo dependiente de IFNa [Poeck, H., et al., 2004. Blood 103(8): p. 3058-64.], y también de la interacción CD70-CD27 [Shaw, J., et al., 2010. Blood 115(15): p. 3051-7]. Dendritic cells (DC) are professional antigen presenting cells (APCs), located at the interface between the innate and adaptive immune response. They are the only APCs capable of stimulating naive T lymphocytes [Villadangos, JA and P. Schnorrer, 2007. Nat Rev Immunol 7 (7): p. 543-555.] And, therefore, have a central role in inducing the autoimmune response. These cells have developed exclusive and probably predominant cross-presentation pathways, which allow them to be restricted to class I molecules of exogenous antigens, after internalization by endocytosis of phagocytosis [Rock, KL, 2003. Nat Immunol 4 (10): p . 941-3]. These pathways are crucial for the stimulation of a specific response of antigens that are not produced in the cytosol of DC, as are those recognized in the case of autoimmune diseases [Segura, E. and JA 2009. Curr Opin Immunol 21 (1): p. 105-10]. DC can mediate very different effects, such as immunity or tolerance, depending on the subpopulation considered and the microenvironment in which they are located [Steinman, RM, et al., 2003. Annu Rev Immunol, 21: p. 685-711]. human DC can be divided into two main subpopulations: conventional myeloid-type (cDC) DCs secrete large amounts of IL-12 and IL-23 after being stimulated with LPS, double-stranded DNA or microbial lipopeptides, thereby inducing a robust Th1 and CD8 effector responses. Phagocytic and presenting cells are prominent, and can be differentiated in vitro from circulating monocytes (MoDC). On the other hand, DC plasmacytoids (pDC) have been classically considered as poor immune system stimulators. However, recent results have shown that peptide-pulsed, or virus-infected, pDCs can effectively stimulate an effector T response [Lui, G., et ai, 2009. PLoS One 4 (9): p. e71 11; Di Pucchio, T., et al., 2008. Nat Immunol 9 (5): p. 551-7.], But also activate the CD4 + or CD8 + regulatory T lymphocytes [Moseman, EA, et al., 2004. J Immunol 173 (7): p. 4433-42]. Although they were supposed to be unable to perform cross-presentation, it has been shown that human circulating pDCs ex vivo cross-presentation of HIV antigens after phagocytosis of infected cells [Hoeffel, G., et al., 2007. Immunity 27 ( 3): p. 481-92]. They produce large amounts of type I and III IFNs in response to viruses, DNA fragments rich in unmethylated CpG motifs, or TLR-7 ligands. The pDCs stimulate the differentiation of plasma cells through IFN type I and IL-6 [Jego, G., et al., 2003. Immunity 19 (2): p. 225-34]. Type I interferons possess other important immunoregulatory functions, including stimulation of T lymphocytes [Bracci, L., et al., 2008. Expert Rev Vaccines 7 (3): p. 373-81]. The ability of any type of DC to induce immunity or tolerance is a consequence of the displacement of the balance between inhibitory or stimulatory factors, determined by the signals of the microenvironment in which the processing and presentation of the antigen occurs. The final phenotype of DC is reflected by the expression of costimulation molecules and cytokine secretion, for which the balance between IL-23, IL-12 and IL-10 is decisive for the polarization of naive T lymphocytes towards a Th1 / Th2 / Th17 / Treg phenotype. Although the interaction between DC and B lymphocytes has been less studied, it has been observed that pDC, but not cDC, can promote differentiation of memory B cells to plasma cells, through an IFNa-dependent mechanism [Poeck , H., et al., 2004. Blood 103 (8): p. 3058-64.], And also from the CD70-CD27 interaction [Shaw, J., et al., 2010. Blood 115 (15): p. 3051-7].
El papel de las DC en el SLE se postuló hace varios años, ya que se detectan altos niveles de IFN de tipo I en pacientes de SLE, y las pDC producen grandes cantidades de estas citoquinas. De hecho, los niveles de pDC en circulación son elevados en pacientes con SLE [Jin, O., et al., 2008. Lupus 17(7): p. 654-62], y también se han encontrado infiltrados de pDC en las lesiones de dichos pacientes [Ko, C.J., et al., 201 1. J Cutan Pathol, 38(1 1): p. 889-92]. Por tanto, las pDC tienen un papel central en la inmunopatogénesis del SLE. The role of DC in SLE was postulated several years ago, since high levels of type I IFNs are detected in SLE patients, and pDCs produce large amounts of these cytokines. In fact, circulating pDC levels are high in patients with SLE [Jin, O., et al., 2008. Lupus 17 (7): p. 654-62], and pDC infiltrates have also been found in the lesions of such patients [Ko, CJ, et al., 201 1. J Cutan Pathol, 38 (1 1): p. 889-92]. Therefore, pDCs have a central role in the immunopathogenesis of SLE.
SIDT1 (SID1 transmembrane family, member 1) es una proteína integral de membrana muy poco estudiada. La mayoría de los datos conocidos sobre SIDT1 hacen referencia a su homólogo en C. elegans, SID1 , una proteína de membrana implicada en el mecanismo de transferencia de iRNA intercelular. SID1 es una proteína con 11 dominios transmembrana, probablemente implicada en el transporte de ácidos nucleicos, concretamente su sobreexpresión aumenta la entrada de moléculas de pequeños RNA de intereferencla (siRNA) en células epiteliales humanas [21-22]. Resultados preliminares apuntan a una localización en el retículo endoplásmico y en vesículas endosomales-lisosomales de SID1. A primera vista parece difícil de concebir cómo SID1 podría estar implicado en la fisiopatología del SLE. Sin embargo, los autores de la presente invención han demostrado que SIDT1 juega un importante papel en el reconocimiento de ácidos nucleicos exógenos y en la iniciación de la respuesta de IFN en las pDC. SIDT1 (SID1 transmembrane family, member 1) is a very poorly studied membrane integral protein. Most of the known data on SIDT1 refers to its counterpart in C. elegans, SID1, a membrane protein involved in the mechanism of intercellular iRNA transfer. SID1 is a protein with 11 transmembrane domains, probably involved in the transport of nucleic acids, specifically its overexpression increases the entry of molecules of small RNA interreferencing (siRNA) into human epithelial cells [21-22]. Preliminary results point to a location in the endoplasmic reticulum and in endosomal-lysosomal vesicles of SID1. At first glance it seems difficult to conceive how SID1 could be involved in the pathophysiology of SLE. However, the authors of the present invention have shown that SIDT1 plays an important role in the recognition of exogenous nucleic acids and in the initiation of the IFN response in pDCs.
Los avances en el descubrimiento de los mecanismos moleculares implicados en enfermedades virales, así como en enfermedades inflamatorias o autoinmunes mediadas por IFN, como la psoriasis o el lupus, permitirían disponer de agentes terapéuticos útiles en tratamiento de estas enfermedades y de métodos para el diagnóstico correcto de las mismas. Advances in the discovery of the molecular mechanisms involved in viral diseases, as well as in IFN-mediated inflammatory or autoimmune diseases, such as psoriasis or lupus, would make it possible to have useful therapeutic agents in the treatment of these diseases and methods for the correct diagnosis from the same.
DESCRIPCIÓN DE LA INVENCIÓN DESCRIPTION OF THE INVENTION
USOS MÉDICOS DE LAS COMPOSICIONES DE LA INVENCIÓN MEDICAL USES OF THE COMPOSITIONS OF THE INVENTION
Los autores de la presente invención han demostrado implicación de SIDT1 en el reconocimiento de ácidos nucleicos exógenos y en la iniciación de la respuesta de IFN en las pDC, y su inhibición disminuye la producción de IFN de tipo I y III. The authors of the present invention have demonstrated involvement of SIDT1 in the recognition of exogenous nucleic acids and in the initiation of the IFN response in the pDCs, and their inhibition decreases the production of type I and III IFNs.
Por tanto, un primer aspecto de la invención se refiere a una composición, de ahora en adelante composición de la invención, que comprende un agente modulador de la actividad de SIDT1 para el tratamiento, mejora o prevención de enfermedades infecciosas, inflamatorias o autoinmunes mediadas por IFN. Therefore, a first aspect of the invention relates to a composition, hereinafter composition of the invention, comprising an agent modulating the activity of SIDT1 for the treatment, improvement or prevention of infectious, inflammatory or autoimmune diseases mediated by IFN
El término " SIDT1 " (SID1 transmembrane family, member 1, también llamado en la literatura SID1 ; SID-1 ; B830021 E24Rik) se localiza en el cromosoma 3 (3q13.2). A no ser que se indique lo contrario, cuando se utiliza en el presente documento el término " SIDT1 " ó " SID1 ", se refiere tanto al gen como a la proteína SIDT1 humana. En el contexto de la presente invención, SIDT1 se define también por una secuencia de nucleótidos o polinucleótido, que constituye la secuencia codificante de la proteína SIDT1 , y que comprendería diversas variantes procedentes de: a) moléculas de ácido nucleico que codifican un polipéptido que comprende la secuencia aminoacídica de la SEQ ID NO: 1 , The term "SIDT1" (SID1 transmembrane family, member 1, also called in the literature SID1; SID-1; B830021 E24Rik) is located on chromosome 3 (3q13.2). Unless otherwise indicated, when the term "SIDT1" or "SID1" is used herein, it refers to both the gene and the human SIDT1 protein. In the context of the present invention, SIDT1 is also defined by a nucleotide or polynucleotide sequence, which constitutes the coding sequence of the SIDT1 protein, and which would comprise various variants from: a) nucleic acid molecules encoding a polypeptide comprising the amino acid sequence of SEQ ID NO: 1,
b) moléculas de ácido nucleico cuya cadena complementarla híbrida con la secuencia polinucleotídica de a),  b) nucleic acid molecules whose chain complement it hybrid with the polynucleotide sequence of a),
c) moléculas de ácido nucleico cuya secuencia difiere de a) y/o b) debido a la degeneración del código genético,  c) nucleic acid molecules whose sequence differs from a) and / or b) due to the degeneracy of the genetic code,
d) moléculas de ácido nucleico que codifican un polipétptido que comprende la secuencia aminoacídica con una identidad de al menos un 80%, un 90%, un 95%, un 98% o un 99% con la SEQ ID NO: 1. en las que el polipéptido codificado por dichos ácidos nucleicos posee la actividad y las características estructurales de la proteína SIDT1. Entre dichas moléculas de ácido nucléico se encuentra la recogida en la secuencia del GenBank (NCBI) NM_017699.2 o SEQ ID NO: 2.  d) nucleic acid molecules encoding a polypeptide comprising the amino acid sequence with an identity of at least 80%, 90%, 95%, 98% or 99% with SEQ ID NO: 1. in that the polypeptide encoded by said nucleic acids possesses the activity and structural characteristics of the SIDT1 protein. Among these nucleic acid molecules is the collection in the GenBank (NCBI) sequence NM_017699.2 or SEQ ID NO: 2.
SEQ ID NO: 1  SEQ ID NO: 1
MRGCLRLALLCALPWLLLAASPGHPAKSPRQPPAPRRDPFDAARGADFDHVYSGWNLSTMRGCLRLALLCALPWLLLAASPGHPAKSPRQPPAPRRDPFDAARGADFDHVYSGWNLST
ENIYSFNYTSQPDQVTAVRVYVNSSSENLNYPVLVWRQQKEVLSWQVPLLFQGLYQRSYENIYSFNYTSQPDQVTAVRVYVNSSSENLNYPVLVWRQQKEVLSWQVPLLFQGLYQRSY
NYQEVSRTLCPSEATNETGPLQQLIFVDVASMAPLGAQYKLLVTKLKHFQLRTNVAFHFTANYQEVSRTLCPSEATNETGPLQQLIFVDVASMAPLGAQYKLLVTKLKHFQLRTNVAFHFTA
SPSQPQYFLYKFPKDVDSVIIKWSEMAYPCSVVSVQNIMCPVYDLDHNVEFNGVYQSMTKSPSQPQYFLYKFPKDVDSVIIKWSEMAYPCSVVSVQNIMCPVYDLDHNVEFNGVYQSMTK
KAAITLQKKDFPGEQFFWFVI KPEDYACGGSFFIQEKENQTWN LQRKKN LEVTIVPSI KESVKAAITLQKKDFPGEQFFWFVI KPEDYACGGSFFIQEKENQTWN LQRKKN LEVTIVPSI KESV
YVKSSLFSVFIFLSFYLGCLLVGFVHYLRFQRKSIDGSFGSNDGSGNMVASHPIAASTPEGSYVKSSLFSVFIFLSFYLGCLLVGFVHYLRFQRKSIDGSFGSNDGSGNMVASHPIAASTPEGS
NYGTIDESSSSPGRQMSSSDGGPPGQSDTDSSVEESDFDTMPDIESDKNIIRTKMFLYLSDNYGTIDESSSSPGRQMSSSDGGPPGQSDTDSSVEESDFDTMPDIESDKNIIRTKMFLYLSD
LSRKDRRIVSKKYKIYFWNIITIAVFYALPVIQLVITYQTWNVTGNQDICYYNFLCAHPLGVLSLSRKDRRIVSKKYKIYFWNIITIAVFYALPVIQLVITYQTWNVTGNQDICYYNFLCAHPLGVLS
AFNNILSNLGHVLLGFLFLLIVLRRDILHRRALEAKDIFAVEYGIPKHFGLFYAMGIALMMEGVAFNNILSNLGHVLLGFLFLLIVLRRDILHRRALEAKDIFAVEYGIPKHFGLFYAMGIALMMEGV
LSACYHVCPNYSNFQFDTSFMYMIAGLCMLKLYQTRHPDINASAYSAYASFAVVIMVTVLGLSACYHVCPNYSNFQFDTSFMYMIAGLCMLKLYQTRHPDINASAYSAYASFAVVIMVTVLG
VVFGKNDVWFWVIFSAIHVLASLALSTQIYYMGRFKIDLGIFRRAAMVFYTDCIQQCSRPLYVVFGKNDVWFWVIFSAIHVLASLALSTQIYYMGRFKIDLGIFRRAAMVFYTDCIQQCSRPLY
MDRMVLLWGNLVNWSFALFGLIYRPRDFASYMLGIFICNLLLYLAFYIIMKLRSSEKVLPVPLMDRMVLLWGNLVNWSFALFGLIYRPRDFASYMLGIFICNLLLYLAFYIIMKLRSSEKVLPVPL
FCIVATAVMWAAALYFFFQNLSSWEGTPAESREKNRECILLDFFDDHDIWHFLSATALFFSFFCIVATAVMWAAALYFFFQNLSSWEGTPAESREKNRECILLDFFDDHDIWHFLSATALFFSF
LVLLTLDDDLDWRRDQI PVF LVLLTLDDDLDWRRDQI PVF
SEQ ID NO: 2 aataaagaaataaaacaggtgtctgaggagaggttctattagagaggggagaatagagagtaattatcgttcagacaggcaa gagttctctgcaggtagcagcctgggaaagcgaactcgcgcaccctcttgggggacgggctcttttgctttctatttcttctcctgcag ccagggtgcacacgtatttgaaacagaccgaatttcctcctcgatgtggcgtcaggttgacttttcgagactagcgggtatttcttttta atgactccaatgcctttttattattgctgttattgaggttgagggagaagagatcggtctaaattctggctgggtaagtggggggattct cggcgatgagaaacgggggacttagaagccggaggaaaatcagcagccccacatctccacttctccagtccgccctactctc cacccgtgacctccagtggagaccccaggcggcagcatcagtatttgatcggcccttcgtcagcacgctgccagccctggccg gctgggttcgccaggcatcacccgctcggctctgaagcggacgcctggccctgcaccgggctttggaaggaccctctctgcgct cgccccctccccagggtggctccgctttcgagcccgggcgcggtgcccaccatgcgcggctgcctgcggctcgcgctgctctgc gcgctgccctggctcctgctggcggcgtcgcccgggcacccggcgaaatcccccaggcagcccccggcaccgcgccgcga ccccttcgacgctgccaggggcgccgatttcgatcatgtctacagcggggtggtgaacctcagcaccgagaacatctactctttc aactacaccagccagcccgaccaggtgacagccgtgagggtgtatgtgaacagttcctctgagaatctcaactacccggtcctt gttgtggttcgccagcagaaagaggtgctgtcctggcaggttcctctgctcttccaaggactataccagaggagctacaactatca agaagtgagccgcaccttatgtccctcagaagcaaccaatgagacgggacccttgcagcaactgatatttgtagatgtcgcatc catggcacccctgggtgctcagtacaaactgctagttaccaagctgaagcacttccagctccggacaaatgttgcctttcactttac tgccagcccctctcaacctcagtattttctatacaagtttcccaaagacgtggactcagttatcattaaagtggtgtctgaaatggctt atccatgttctgttgtctcagtccagaatatcatgtgcccggtgtatgatctcgaccacaatgtggaatttaatggtgtctatcagtccat gaccaagaaagctgccatcacgctacagaagaaggattttccaggcgagcagttcttcgtggtatttgtgataaagcctgaagat tatgcctgtggaggatctttcttcatccaggaaaaggaaaaccagacctggaatctacagcgaaaaaagaaccttgaagtgac cattgtcccttccattaaagaatctgtttatgtgaaatccagtcttttcagtgtcttcatcttcctgtccttctacttgggatgccttcttgttgg gtttgttcattatctgaggtttcagagaaaatccattgatggaagctttgggtccaatgatggctctggaaatatggtggcatctcatcc cattgctgccagcacacccgaagggagcaattatgggacaatagatgagtcaagctccagtcctggaaggcagatgtcctcct ccgatggtgggccaccgggccagtcagacacagacagctccgtggaggagagcgacttcgacaccatgccagacattgag agtgataaaaacatcatccggaccaagatgttcctttacctgtcagatttgtccaggaaggaccggagaattgtcagcaaaaaat ataaaatttatttttggaacatcatcaccattgctgtgttttacgcgctgcccgtgatccagctggtcattacctatcagacagtggtaa atgtcactggcaaccaggacatctgttactacaacttcctctgtgctcaccccttgggcgtcctgagtgccttcaacaacattctcag caatctgggccacgtgcttctgggcttcctcttcctgctgatagtcttgcgccgcgacatcctccatcggagagccctggaagccaa ggacatctttgctgtggagtacgggattcccaaacactttggtctcttctacgctatgggcattgcattgatgatggaaggggtgctc agtgcttgctaccatgtctgccctaattattccaacttccaattcgacacctccttcatgtacatgatcgctggcctgtgcatgctgaag ctctatcagacccgccacccagacatcaatgccagcgcctactctgcctatgcctcctttgctgtggtcatcatggtcaccgtccttg gagtggtgtttggaaaaaatgacgtatggttctgggtcatcttctctgcaatccacgttctggcctcgctagccctcagcacccagat atattatatgggtcgtttcaagatagatttgggaattttccggcgggctgccatggtgttctacacagactgtatccagcagtgtagcc gacctctatatatggatagaatggtgttgctggttgtggggaatctggttaactggtccttcgccctctttggattgatataccgcccca gggactttgcttcctacatgctgggcatcttcatctgtaaccttttgctgtacctggccttttacatcatcatgaagctccgcagctctga aaaggtcctcccagtcccgctcttctgcatcgtggccaccgctgtgatgtgggctgccgccctatattttttcttccagaatctcagca gctgggagggaactccggccgaatcccgggagaagaaccgcgagtgcattctgctggatttcttcgatgaccatgacatctggc acttcctctctgctactgctctgtttttctcattcttggttttgttaactttggatgatgaccttgatgtggttcggagagaccagatccctgtc ttctgaacctccaacattaagagaggggagggagcgatcaatcttggtgctgtttcacaaaaattacagtgaccacagcaaagt aaccactgccagatgctccactcaccctctgtagagccaactctgcattcacacaggaaggagaggggctgcgggagatttaa acctgcaagaaaggaggcagaaggggagccatgttttgaggacagacgcaaacctgaggagctgagaaacacttgctcctt ccatctgcagctttgggagtgcaacagggataggcactgcatccaagtcaactcaccatcttggggtccctcccaccctcacgg agacttgccagcaatggcagaatgctgctgcacacttccctccagttgtcaccctgcccagaaaggccagcagcttggacttcct gcccagaaactgtgttggcccccttcacacctctgcaacacctgctgctccagcaagaggatgtgattctttagaatatggcgggg aggtgaccccaggccctgccctactgggatagatgttttaatggcaccagctagtcacctcccagaagaaactctgtatatttccc ccaggtttctgatgccatcagaagggctcaggagtggggtttgtcacacattcctcttaacaagtaactgtcactgggaccgagtc ctgggtgcttacatattccttcgtgtcttcatctcactgacctgtgtggacctcatcactctgactctgccttcttggaaaggccctgtca ctccacagatgtctggccagcttcaaggcagaaggaaaaacaggaaaagctcttttaacagcagcaggaacaagagaaatg actaaccatactaaaagactggtaacagcagcagcagccagacaggcctcaccttaaggacttgggctgccagagcaaattc agcagagcttatttggcctcccattcacacagctcagttctgtgcccacatcacctttggggaagaaatcagcattctaatcaggg acactacttcaggagtcctccacagcgagtccgtcatctgtcactttatgtagatcagggttctagacttcttccctgaggttctcaga agcagctctcaggatgaacgtattgtcctcttcccctcttcttgcaaagtgcacagctaatctaatgttgtctctcggttgcacctgaca ttctctccccagtaaggtgttggcaagctcagcatctgggttccactctcacactgtctggcagctctgtgtctgagaagttctacattg accaggcccccttgttgcctggagtatgacgtaatcagaaaatagacgtataaatgtgcacatgcgtatgtatttgcttgtgaaatta aagtcacctcttgcctctgctttcctgatcattcgttagagaaatggatcaggcatttttttaaattattattctttctctaaactatttgcatt gtgttcaaaaacccattttagaagtttgaacagcaagcttttcctgattttaaaaacacaaagttgctttcaatgaaatattttgtgatttt tttaaagtccccaaatgtgtacttagccttctgttattccttattctttaagcagtgttggcttccattgaccatatgaaggccaccaatta aatggttgtgttaatccaacatgtaaaaaactttttggcagggcacagtggctcacgcctgtaatcccaacactttgggaggctga ggcaggaggatcacttgagcccaggagattgacgccgcagtgaactatgattgtgcccctgcactccagcctggatgacagag tgagaccccatctcttaaaaaataaaaaaaaataaaaatttgaacctgtttcaaaaaaaaaaaaaaaaaaaa SEQ ID NO: 2 gagttctctgcaggtagcagcctgggaaagcgaactcgcgcaccctcttgggggacgggctcttttgctttctatttcttctcctgcag aataaagaaataaaacaggtgtctgaggagaggttctattagagaggggagaatagagagtaattatcgttcagacaggcaa ccagggtgcacacgtatttgaaacagaccgaatttcctcctcgatgtggcgtcaggttgacttttcgagactagcgggtatttcttttta atgactccaatgcctttttattattgctgttattgaggttgagggagaagagatcggtctaaattctggctgggtaagtggggggattct cggcgatgagaaacgggggacttagaagccggaggaaaatcagcagccccacatctccacttctccagtccgccctactctc cacccgtgacctccagtggagaccccaggcggcagcatcagtatttgatcggcccttcgtcagcacgctgccagccctggccg gctgggttcgccaggcatcacccgctcggctctgaagcggacgcctggccctgcaccgggctttggaaggaccctctctgcgct cgccccctccccagggtggctccgctttcgagcccgggcgcggtgcccaccatgcgcggctgcctgcggctcgcgctgctctgc gcgctgccctggctcctgctggcggcgtcgcccgggcacccggcgaaatcccccaggcagcccccggcaccgcgccgcga ccccttcgacgctgccaggggcgccgatttcgatcatgtctacagcggggtggtgaacctcagcaccgagaacatctactctttc aactacaccagccagcccgaccaggtgacagccgtgagggtgtatgtgaacagttcctctgagaatctcaactacccggtcctt gttgtggttcgccagcagaaagaggtgctgtcctggcaggttcctctgctcttccaaggactataccagaggagctacaactatca agaagtgagccgcaccttatgtccctcagaagcaaccaatgagacgggacccttgcagcaactgatatttgtagatgtcgcatc catggcacccctgggtgctcagtacaaactgctagttaccaagctgaagcacttccagctccggacaaatgttgcctttcactttac tgccagcccctctcaacctcagtattttctatacaagtttcccaaagacgtggactcagt tatcattaaagtggtgtctgaaatggctt atccatgttctgttgtctcagtccagaatatcatgtgcccggtgtatgatctcgaccacaatgtggaatttaatggtgtctatcagtccat gaccaagaaagctgccatcacgctacagaagaaggattttccaggcgagcagttcttcgtggtatttgtgataaagcctgaagat tatgcctgtggaggatctttcttcatccaggaaaaggaaaaccagacctggaatctacagcgaaaaaagaaccttgaagtgac cattgtcccttccattaaagaatctgtttatgtgaaatccagtcttttcagtgtcttcatcttcctgtccttctacttgggatgccttcttgttgg gtttgttcattatctgaggtttcagagaaaatccattgatggaagctttgggtccaatgatggctctggaaatatggtggcatctcatcc cattgctgccagcacacccgaagggagcaattatgggacaatagatgagtcaagctccagtcctggaaggcagatgtcctcct ccgatggtgggccaccgggccagtcagacacagacagctccgtggaggagagcgacttcgacaccatgccagacattgag agtgataaaaacatcatccggaccaagatgttcctttacctgtcagatttgtccaggaaggaccggagaattgtcagcaaaaaat ataaaatttatttttggaacatcatcaccattgctgtgttttacgcgctgcccgtgatccagctggtcattacctatcagacagtggtaa atgtcactggcaaccaggacatctgttactacaacttcctctgtgctcaccccttgggcgtcctgagtgccttcaacaacattctcag caatctgggccacgtgcttctgggcttcctcttcctgctgatagtcttgcgccgcgacatcctccatcggagagccctggaagccaa g gacatctttgctgtggagtacgggattcccaaacactttggtctcttctacgctatgggcattgcattgatgatggaaggggtgctc agtgcttgctaccatgtctgccctaattattccaacttccaattcgacacctccttcatgtacatgatcgctggcctgtgcatgctgaag ctctatcagacccgccacccagacatcaatgccagcgcctactctgcctatgcctcctttgctgtggtcatcatggtcaccgtccttg gagtggtgtttggaaaaaatgacgtatggttctgggtcatcttctctgcaatccacgttctggcctcgctagccctcagcacccagat atattatatgggtcgtttcaagatagatttgggaattttccggcgggctgccatggtgttctacacagactgtatccagcagtgtagcc gacctctatatatggatagaatggtgttgctggttgtggggaatctggttaactggtccttcgccctctttggattgatataccgcccca aaaggtcctcccagtcccgctcttctgcatcgtggccaccgctgtgatgtgggctgccgccctatattttttcttccagaatctcagca gggactttgcttcctacatgctgggcatcttcatctgtaaccttttgctgtacctggccttttacatcatcatgaagctccgcagctctga gctgggagggaactccggccgaatcccgggagaagaaccgcgagtgcattctgctggatttcttcgatgaccatgacatctggc acttcctctctgctactgctctgtttttctcattcttggttttgttaactttggatgatgaccttgatgtggttcggagagaccagatccctgtc ttctgaacctccaacattaagagaggggagggagcgatcaatcttggtgctgtttcacaaaaattacagtgaccacagcaaagt aaccactgccagat gctccactcaccctctgtagagccaactctgcattcacacaggaaggagaggggctgcgggagatttaa acctgcaagaaaggaggcagaaggggagccatgttttgaggacagacgcaaacctgaggagctgagaaacacttgctcctt ccatctgcagctttgggagtgcaacagggataggcactgcatccaagtcaactcaccatcttggggtccctcccaccctcacgg agacttgccagcaatggcagaatgctgctgcacacttccctccagttgtcaccctgcccagaaaggccagcagcttggacttcct gcccagaaactgtgttggcccccttcacacctctgcaacacctgctgctccagcaagaggatgtgattctttagaatatggcgggg aggtgaccccaggccctgccctactgggatagatgttttaatggcaccagctagtcacctcccagaagaaactctgtatatttccc ccaggtttctgatgccatcagaagggctcaggagtggggtttgtcacacattcctcttaacaagtaactgtcactgggaccgagtc ctgggtgcttacatattccttcgtgtcttcatctcactgacctgtgtggacctcatcactctgactctgccttcttggaaaggccctgtca ctccacagatgtctggccagcttcaaggcagaaggaaaaacaggaaaagctcttttaacagcagcaggaacaagagaaatg actaaccatactaaaagactggtaacagcagcagcagccagacaggcctcaccttaaggacttgggctgccagagcaaattc agcagagcttatttggcctcccattcacacagctcagttctgtgcccacatcacctttggggaagaaatcagcattctaatcaggg acactacttcaggagtcctccacagcgagtccgtcatctgtcactttatgtagatcagggttctagacttcttccctgaggttctcaga agcagctctcaggatgaacgtattgtcctcttcccctcttcttgcaaagtgcacagctaatctaatgttgtctctcggttgcacctgaca ttctctccccagtaaggtgttggcaagctcagcatctgggttccactctcacactgtctggcagctctgtgtctgagaagttctacattg accaggcccccttgttgcctggagtatgacgtaatcagaaaatagacgtataaatgtgcacatgcgtatgtatttgcttgtgaaatta aagtcacctcttgcctctgctttcctgatcattc gttagagaaatggatcaggcatttttttaaattattattctttctctaaactatttgcatt gtgttcaaaaacccattttagaagtttgaacagcaagcttttcctgattttaaaaacacaaagttgctttcaatgaaatattttgtgatttt tttaaagtccccaaatgtgtacttagccttctgttattccttattctttaagcagtgttggcttccattgaccatatgaaggccaccaatta aatggttgtgttaatccaacatgtaaaaaactttttggcagggcacagtggctcacgcctgtaatcccaacactttgggaggctga ggcaggaggatcacttgagcccaggagattgacgccgcagtgaactatgattgtgcccctgcactccagcctggatgacagag tgagaccccatctcttaaaaaataaaaaaaaataaaaatttgaacctgtttcaaaaaaaaaaaaaaaaaaaa
El término "que modula la actividad" como se usa aquí, se refiere tanto a que inhibe (disminuye) o estimula (incrementa) el nivel de actividad de la proteína SIDT1 en una célula. La actividad de SIDT1 puede ser modulada por la modificación de los niveles y/o de la actividad de la proteína SIDT1 , o por la modificación de los niveles a los que se transcribe el gen SIDT1 tal que los niveles de actividad de la proteína SIDT1 en la célula es modulada. Los agentes moduladores pueden ser también agonistas (sustancias que son capaces de unirse a un receptor y provocar una respuesta en la célula, en concreto un aumento de la actividad de SIDT1), como antagonistas (sustancias que no solamente no activan el receptor, sino que en realidad bloquea su activación por los agonistas). En el contexto de la presente invención, la inhibición es la forma preferida de modulación. The term "modulating activity" as used herein, refers both to inhibiting (decreasing) or stimulating (increasing) the level of activity of the SIDT1 protein in a cell. The activity of SIDT1 can be modulated by the modification of the levels and / or activity of the SIDT1 protein, or by the modification of the levels to which the SIDT1 gene is transcribed such that the levels of activity of the SIDT1 protein in The cell is modulated. Modulating agents can also be agonists (substances that are capable of binding to a receptor and eliciting a response in the cell, specifically an increase in SIDT1 activity), as antagonists (substances that not only do not activate the receptor, but also actually blocks its activation by agonists). In the context of the present invention, inhibition is the preferred form of modulation.
En una realización preferida de este aspecto de la invención, los agentes moduladores comprendidos en la composición de la invención se seleccionan de una lista que comprende: a) una molécula orgánica, b) una molécula de ARN, c) un oligonucleótido antisentido, d) un anticuerpo, o e) una ribozima F) Una nucleasa con capacidad de editar el ADN cromosómico (ZFN, Talen, CRISPR). In a preferred embodiment of this aspect of the invention, the modulating agents comprised in the composition of the invention are selected from a list comprising: a) an organic molecule, b) an RNA molecule, c) an antisense oligonucleotide, d) an antibody, or e) a ribozyme F) A nuclease capable of editing chromosomal DNA (ZFN, Talen, CRISPR).
Un experto en la materia podría preparar moléculas orgánicas que pueden unirse específicamente a la SIDT1 sin unirse a otros polipéptidos o proteínas, las moléculas orgánicas tendrán preferiblemente un peso de 100 a 20.000 daltons, más preferiblemente 500 a 15.000 daltons, y más preferiblemente 1000 a 10.000 daltons. Librerías de moléculas orgánicas se encuentran disponibles comercialmente. La vía de administración puede ser, sin limitarse a estas, intraperitoneal, intratecal, intravenosa, intramuscular, subcutánea, intraventricular, oral, enteral, parenteral, intranasal o dérmica. Recientemente, con el desarrollo de la tecnología antisentido, secuencias de nucleótidos específicamente complementarios a una determinada secuencia de ADN o ARN, podrían formar complejos y bloquear la transcripción o traducción. Así, con el progreso del silenciamiento génico post-transcripcional, y en particular del ARN de interferencia (RNA interferente o RNAi), se han desarrollado herramientas que permiten la inhibición específica de la expresión de un gen. La inhibición de la expresión de la proteína SIDT1 constituiría por ende la inhibición de su actividad biológica, y en concreto, de la actividad que está contribuyendo al agravamiento de las características inflamatorias o autoinmunes mediadas por IFN. One skilled in the art could prepare organic molecules that can specifically bind SIDT1 without binding to other polypeptides or proteins, the organic molecules will preferably have a weight of 100 to 20,000 daltons, more preferably 500 to 15,000 daltons, and more preferably 1000 to 10,000 daltons. Bookstores of organic molecules are commercially available. The route of administration may be, without limitation, intraperitoneal, intrathecal, intravenous, intramuscular, subcutaneous, intraventricular, oral, enteral, parenteral, intranasal or dermal. Recently, with the development of antisense technology, nucleotide sequences specifically complementary to a particular DNA or RNA sequence could form complexes and block transcription or translation. Thus, with the progress of post-transcriptional gene silencing, and in particular of interfering RNA (interfering RNA or RNAi), tools have been developed that allow specific inhibition of gene expression. The inhibition of the expression of the SIDT1 protein would therefore constitute the inhibition of its biological activity, and in particular, of the activity that is contributing to the aggravation of the inflammatory or autoimmune characteristics mediated by IFN.
Por "polinucleótidos antisentido" se entienden cadenas de ribonucleótidos o desoxirribonucleóitidos que pueden inhibir la producción de la proteína SIDT1 por uno de estos tres mecanismos: By "antisense polynucleotides" are meant ribonucleotide or deoxyribonucleotide chains that can inhibit the production of the SIDT1 protein by one of these three mechanisms:
1- Interfiriendo la transcripción, al hibridar en el gen estructural o en una región regulatoria del gen que codifica para SIDT1. Puesto que la transcripción o expresión es bloqueada de manera efectiva por la hibridación del oligonucleótido antisentido con el ADN, disminuye la producción de SIDT1. 2- La unión del oligonucleótido antisentido en el citoplasma con el ARNm, interfiriendo con la formación de la construcción de traducción propiamente dicha, inhibiendo la traducción de ARNm a la proteína. 1- Interfering transcription by hybridizing in the structural gene or in a regulatory region of the gene encoding SIDT1. Since transcription or expression is effectively blocked by hybridization of the antisense oligonucleotide with DNA, the production of SIDT1 decreases. 2- The binding of the antisense oligonucleotide in the cytoplasm with the mRNA, interfering with the formation of the translation construct itself, inhibiting the translation of mRNA into the protein.
3- La formación de un ARNm - antisentido dúplex que permite una rápida degradación del ARNm dúplex por ARNasas (como ARNasa H). Esto da lugar a una menor producción de SIDT1. 3- The formation of a duplex antisense mRNA that allows rapid degradation of the duplex mRNA by RNases (such as RNase H). This results in a lower production of SIDT1.
Oligonucleótidos antisentido capaces de modular la actividad de la SIDT1 se pueden sintetizar fácilmente a partir de la secuencia conocida SEQ ID NO: 2 aplicando el conocimiento del estado de la técnica. Por ejemplo, y sin limitarnos, podría ser una secuencia de ribonucleótidos o ARN que pertenece al denominado siRNA (small interfering RNA), ARN pequeño de interferencia o ARN de silenciamiento, capaz de inhibir la expresión genética de la proteína SIDT1. En el contexto de la presente memoria se entiende como "siRNA" (small interfering RNA ó ARN pequeño de interferencia) una clase de ARN de doble cadena de 19 a 25 nucleótidos de largo, y más preferentemente entre 21 y 23 nucleótidos, que está involucrado en la ruta de la interferencia de ARN, donde el siRNA interfiere la expresión de un gen específico. En la presente invención, este gen específico es el SIDT1. También podría ser cualquier siRNA capaz de hibridar una molécula de ácido nucleico que codifique la proteína SIDT1 humana que se recoge en la SEQ ID NO: 1. También podrían ser una construcción de ARN que al menos contenga una cualquiera de las secuencias de nucleótidos posibles de siRNA capaces de inhibir la expresión de SIDT1 , y sin perjuicio de que adicionalmente formen parte de la presente invención cualquiera de las secuencias y construcciones de RNA de la invención anteriormente descritas que sean objeto de modificaciones, preferentemente químicas, que conduzcan a una mayor estabilidad frente a la acción de ribonucleasas y con ello a una mayor eficiencia. Sin que dichas modificaciones supongan la alteración de su mecanismo de acción, que es la unión específica al complejo RISC (RNA-induced silencing complex), activándolo y manifestando una actividad helicasa que separa las dos hebras dejando solo la hebra antisentido asociada al complejo. El complejo ribonucleoprotéico resultante se une al mRNA diana (ARN mensajero del SIDT1 , que se recoge en la SEQ ID NO: 2). Si la complementariedad no es perfecta, RISC queda asociado al mensajero y se atenúa la traducción. Pero si es perfecta, RISC actúa como RNasa, cortando al mensajero y quedando libre para repetir el proceso. Adicionalmente resulta evidente para un experto en la materia que una gran cantidad de polinucleótidos de mARN pueden traducirse a SIDT1 como consecuencia, por ejemplo, de que el código genético es degenerado. Cualquier siRNA capaz de inhibir la traducción de estos mARN también forma parte de la invención. Antisense oligonucleotides capable of modulating the activity of SIDT1 can be easily synthesized from the known sequence SEQ ID NO: 2 by applying knowledge of the state of the art. For example, and without limiting ourselves, it could be a sequence of ribonucleotides or RNA that belongs to the so-called siRNA (small interfering RNA), small interfering RNA or silencing RNA, capable of inhibiting the genetic expression of the SIDT1 protein. In the context of the present specification, "siRNA" (small interfering RNA or small interfering RNA) is understood to be a class of double stranded RNA 19 to 25 nucleotides long, and more preferably between 21 and 23 nucleotides, which is involved in the route of RNA interference, where siRNA interferes with the expression of a specific gene. In the present invention, this specific gene is SIDT1. It could also be any siRNA capable of hybridizing a nucleic acid molecule encoding the human SIDT1 protein that is collected in SEQ ID NO: 1. They could also be an RNA construct that contains at least any one of the possible nucleotide sequences of siRNA capable of inhibiting the expression of SIDT1, and without prejudice to the addition of any of the RNA sequences and constructions of the invention described above that are subject to modifications, preferably chemical, leading to greater stability against the action of ribonucleases. and with it to greater efficiency. Without these modifications involving the alteration of its mechanism of action, which is the specific binding to the RISC complex (RNA-induced silencing complex), activating it and manifesting a helicase activity that separates the two strands leaving only the antisense strand associated with the complex. The resulting ribonucleoproteic complex binds to the target mRNA (messenger RNA of SIDT1, which is collected in SEQ ID NO: 2). If the complementarity is not perfect, RISC is associated with the messenger and the translation is attenuated. But if it is perfect, RISC acts as RNasa, cutting the messenger and being free to repeat the process. Additionally, it is apparent to one skilled in the art that a large number of mRNA polynucleotides can be translated into SIDT1 as a consequence, for example, that the genetic code is degenerated. Any siRNA capable of inhibiting the translation of these mRNAs is also part of the invention.
La preparación de la secuencia de siRNA de la invención o de la construcción de RNA de la invención sería evidente para un experto en la materia, y se podría llevar a cabo por síntesis química, lo cual permite además la incorporación de modificaciones químicas tanto en los distintos nucleótidos del producto como la incorporación de otros compuestos químicos en cualquiera de los extremos. Por otro lado, la síntesis también podría realizarse enzimáticamente utilizando cualquiera de las RNA polimerasas disponibles. La síntesis enzimática también permite alguna modificación química de los productos o RNAs inhibidores. The preparation of the siRNA sequence of the invention or of the RNA construct of the invention would be apparent to one skilled in the art, and could be carried out by chemical synthesis, which also allows the incorporation of chemical modifications in both different nucleotides of the product such as the incorporation of other chemical compounds at any of the ends. On the other hand, the synthesis could also be carried out enzymatically using any of the available RNA polymerases. Enzymatic synthesis also allows some chemical modification of inhibitor products or RNAs.
El diseño de la secuencia de nucleótidos del siRNA de la invención también sería evidente para un experto en la materia. Así, se podría realizar mediante un diseño aleatorio en el que se seleccionen 19-25 bases del ARNm diana sin tener en cuenta la secuencia o la información posicional que tiene en el transcrito. Otra alternativa no limitativa de la presente invención sería el diseño convencional mediante parámetros simples desarrollados por los pioneros de la técnica (Calipel, A. et al., 2003. J Biol Chem. 278(43): 42409-42418) completados con un análisis BLAST de nucleótidos. Otra posibilidad podría ser un diseño racional, en el que se emplee un procedimiento informático dirigido a identificar las dianas óptimas de siRNA en un ARNm. Las secuencias diana se analizan en grupos de 19 nucleótidos a la vez y se identifican las que tienen mejores características en función de un algoritmo que incorpora un gran número de parámetros termodinámicos y de secuencia. También podría formar parte de la composición de la invención una construcción genética de ADN, la cual dirigiría la transcripción in vitro o intracelular de la secuencia siRNA o construcción de ARN de la invención, y que comprende, al menos, uno de los siguientes tipos de secuencias: a) secuencia de nucleótidos de ADN, preferentemente de doble cadena, que comprende, al menos, la secuencia codificante del siRNA de la invención o de la construcción de ARN de la invención para su transcripción, o, b) secuencia de nucleótidos de ADN, preferentemente de doble cadena, correspondiente a un sistema o vector de expresión génica que comprende la secuencia codificante de la secuencia de ARN de la invención operativamente enlazada con, al menos, un promotor que dirija la transcripción de dicha secuencia de nucleótidos de interés, y con otras secuencias necesarias o apropiadas para la transcripción y su regulación adecuada en tiempo y lugar, por ejemplo, señales de inicio y terminación, sitios de corte, señal de poliadenilación, origen de replicación, activadores transcripcionales (enhancers), silenciadores transcripcionales (silencers), etc. para su uso en aquellos contextos patológicos en los que se encuentre alterados los niveles de IFN, y preferiblemente, IFN de tipo I y III. The nucleotide sequence design of the siRNA of the invention would also be apparent to one skilled in the art. Thus, it could be done through a random design in which 19-25 bases of the target mRNA are selected without taking into account the sequence or positional information it has in the transcript. Another non-limiting alternative of the present invention would be the conventional design by simple parameters developed by the pioneers of the art (Calipel, A. et al., 2003. J Biol Chem. 278 (43): 42409-42418) completed with an analysis BLAST nucleotide. Another possibility could be a rational design, in which a computer procedure is used to identify the optimal siRNA targets in an mRNA. The target sequences are analyzed in groups of 19 nucleotides at a time and those with the best characteristics are identified based on an algorithm that incorporates a large number of thermodynamic and sequence parameters. A genetic DNA construct could also be part of the composition of the invention, which would direct the in vitro or intracellular transcription of the siRNA sequence or RNA construct of the invention, and comprising at least one of the following types of sequences: a) DNA nucleotide sequence, preferably double stranded, comprising at least the sequence encoding the siRNA of the invention or the RNA construct of the invention for transcription, or, b) nucleotide sequence of DNA, preferably double stranded, corresponding to a gene expression system or vector comprising the sequence coding for the RNA sequence of the invention operably linked with at least one promoter that directs the transcription of said nucleotide sequence of interest, and with other sequences necessary or appropriate for transcription and its appropriate regulation in time and place, for example, start signals and termination, cutting sites, polyadenylation signal, origin of replication, transcriptional activators (enhancers), transcriptional silencers (silencers), etc. for use in those pathological contexts in which IFN levels are altered, and preferably, type I and III IFNs.
SIDT1 está contribuyendo al agravamiento de las características inflamatorias y/o autoinmunes y a la aparición de la enfermedad. Múltiples de estas construcciones, sistemas o vectores de expresión pueden ser obtenidos por métodos convencionales conocidos por los expertos en la materia (Sambrook et al. 2001. Molecular Cloning: A laboratory Manual. Coid Spring Harbor laboratory Press, New York) SIDT1 is contributing to the worsening of inflammatory and / or autoimmune characteristics and to the onset of the disease. Multiple of these constructs, systems or expression vectors can be obtained by conventional methods known to those skilled in the art (Sambrook et al. 2001. Molecular Cloning: A laboratory Manual. Coid Spring Harbor laboratory Press, New York)
Las composiciones de la presente invención permiten la transfección del siRNA de la invención al interior de una célula, in vivo o in vitro. La transfección se podría llevar a cabo, pero sin limitarnos a, transfección directa o vectores que faciliten el acceso del siRNA al interior de la célula. Así, ejemplos de estos vectores son, sin limitarse a, retrovirus, lentivirus, adenovirus, virus adeno-asociados, virus del Herpes simplex, plásmidos de DNA no virales, liposomas catiónicos y conjugados moleculares. Así, por ejemplo, los siRNA de la presente invención, así como ARN o ADN precursores de estos siRNA, pueden conjugarse con péptidos de liberación u otros compuestos para favorecer el transporte de estos siRNA al interior de la célula. The compositions of the present invention allow the transfection of the siRNA of the invention into a cell, in vivo or in vitro. Transfection could be carried out, but not limited to, direct transfection or vectors that facilitate the access of siRNA into the cell. Thus, examples of these vectors are, without limitation, retroviruses, lentiviruses, adenoviruses, adeno-associated viruses, Herpes simplex viruses, non-viral DNA plasmids, cationic liposomes and molecular conjugates. Thus, for example, the siRNAs of the present invention, as well as RNA or DNA precursors of these siRNAs, can be conjugated with release peptides or other compounds to favor the transport of these siRNAs into the cell.
También forman parte de la invención nucleasas diseñadas para bloquear o activar la expresión del gen. Nucleases designed to block or activate gene expression are also part of the invention.
El término "anticuerpo" tal como se emplea en esta memoria, se refiere a moléculas de inmunoglobulinas y porciones inmunológicamente activas de moléculas de inmunoglobulinas, es decir, moléculas que contienen un sitio de fijación de antígeno que se une específicamente (inmunorreacciona con) la proteína SIDT1. Ejemplos de porciones de moléculas de inmunoglobulinas inmunológicamente activas, incluyen fragmentos F(ab) y F(ab')2 que pueden ser generados tratando el anticuerpo con una enzima tal como la pepsina. Puede ser un anticuerpo monoclonal o policlonal. The term "antibody" as used herein refers to immunoglobulin molecules and immunologically active portions of immunoglobulin molecules, that is, molecules that contain an antigen binding site that specifically binds (immunoreacts with) the protein. SIDT1. Examples of portions of immunologically active immunoglobulin molecules, include F (ab) and F (ab ') 2 fragments that can be generated by treating the antibody with an enzyme such as pepsin. It can be a monoclonal or polyclonal antibody.
Los anticuerpos capaces de unirse a la proteína SIDT1 pueden ser empleados para inhibir la actividad de dicha proteína. Muchos de tales anticuerpos están disponibles comercialmente. Los anticuerpos, o fragmentos de los mismos, podrían ser capaces de inhibir la actividad de la proteína SIDT1 que contribuye a la adquisición de las características propias de las enfermedades inflamatorias y/o autoinmunes. Los anticuerpos pueden ser policlonales (incluyen típicamente anticuerpos distintos dirigidos contra determinantes o epitopos distintos) o monoclonales (dirigidos contra un único determinante en el antígeno). El anticuerpo monoclonal puede ser alterado bioquímicamente, por manipulación genética, o puede ser sintético, careciendo, posiblemente, el anticuerpo en su totalidad o en partes, de porciones que no son necesarias para el reconocimiento de la SIDT1 y estando sustituidas por otras que comunican al anticuerpo propiedades ventajosas adicionales. El anticuerpo puede ser también recombinante, quimérico, humanizado, sintético o una combinación de cualquiera de los anteriores. Antibodies capable of binding to the SIDT1 protein can be used to inhibit the activity of said protein. Many such antibodies are commercially available. The antibodies, or fragments thereof, may be able to inhibit the activity of the SIDT1 protein that contributes to the acquisition of the characteristics of inflammatory and / or autoimmune diseases. The antibodies can be polyclonal (typically include different antibodies directed against different determinants or epitopes) or monoclonal (directed against a single determinant in the antigen). The monoclonal antibody can be biochemically altered, by genetic manipulation, or it can be synthetic, possibly lacking the antibody in whole or in parts, of portions that are not necessary for the recognition of SIDT1 and being replaced by others that communicate to the antibody additional advantageous properties. The antibody can also be recombinant, chimeric, humanized, synthetic or a combination of any of the foregoing.
Un "anticuerpo o polipéptido recombinante" (rAC) es uno que ha sido producido en una célula hospedadora que ha sido transformada o transfectada con el ácido nucleico codificante del polipéptido, o produce el polipéptido como resultado de la recombinación homologa. A "recombinant antibody or polypeptide" (rAC) is one that has been produced in a host cell that has been transformed or transfected with the nucleic acid encoding the polypeptide, or produces the polypeptide as a result of homologous recombination.
Estos rAc se pueden expresar y dirigir hacia subcompartimentos celulares específicos cuando se les incorpora las secuencias apropiadas para el tráfico intracelular. Estos anticuerpos se denominan intrabodies, y han demostrado su eficacia no sólo para desviar proteínas de su compartimento habitual o bloquear interacciones entre proteínas implicadas en vías de señalización, sino también para activar proteínas intracelulares. These rAc can be expressed and directed to specific cellular sub-compartments when the appropriate sequences for intracellular traffic are incorporated. These antibodies are called intrabodies, and have proven effective not only to divert proteins from their usual compartment or block interactions between proteins involved in signaling pathways, but also to activate intracellular proteins.
También forman parte de la invención las construcciones genéticas de DNA capaces de transcribirse a un péptido, anticuerpo o fragmento de anticuerpo, para su uso en el tratamiento, mejora o prevención de enfermedades infecciosas, inflamatorias o autoinmunes mediadas por IFN. Dicha construcción genética de DNA dirigiría la transcripción in vitro o intracelular de la secuencia del anticuerpo o fragmento del mismo, y comprende, al menos, uno de los siguientes tipos de secuencias: a) secuencia de nucleótidos de DNA, preferentemente de doble cadena, que comprende, al menos, la secuencia codificante del anticuerpo de la invención o del fragmento de anticuerpo de la invención para su transcripción in vitro, o intracelular, b) secuencia de nucleótidos de DNA, preferentemente de doble cadena, correspondiente a un sistema o vector de expresión génica que comprende la secuencia codificante de la secuencia de anticuerpo o fragmento de anticuerpo de la invención operativamente enlazada con, al menos, un promotor que dirija la transcripción de dicha secuencia de nucleótidos de interés, y con otras secuencias necesarias o apropiadas para la transcripción y su regulación adecuada en tiempo y lugar, por ejemplo, señales de inicio y terminación, sitios de corte, señal de poliadenilación, origen de replicación, activadores transcripcionales (enhancers), silenciadores transcripcionales (silencers), etc. para su uso en aquellos contextos patológicos que transcurren con liberación de IFN, preferiblemente de tipo I y III. Un "ribozima" tal y como se entiende en la presente invención, se refiere a un polinucleótido catalítico (típicamente RNA), que puede construirse para reconocer específicamente, por hibridación, un mRNA y fragmentarlo o eliminar su expresión. Las ribozimas pueden introducirse en la célula como moléculas de RNA catalíticas o como construcciones genéticas que se expresan a moléculas catalíticas de RNA. El término "enfermedad autoinmune" se refiere a una afección en un sujeto caracterizada por una lesión celular, tisular y/o de los órganos causada por una reacción inmunológica del sujeto a sus propias células, tejidos y/u órganos. Ejemplos ilustrativos , no limitativos de enfermedades autoinmunes incluyen la alopecia areata, la espondilitis anquilosante, síndrome antifosfolípidos, enfermedad autoinmune de Addison, enfermedades autoinmunes de la glándula suprarrenal, la anemia hemolítica autoinmune, hepatitis autoinmune, ooforitis autoinmune y orquitis, trombocitopenia autoinmune, enfermedad de Behcet, penfigoide bulloso, cardiomiopatía, enfermedad celíaca , la dermatitis , la fatiga crónica , síndrome de disfunción inmune ( SFC ), la polineuropatía desmielinizante inflamatoria crónica , síndrome de Churg - Strauss, penfigoide cicatricial, síndrome de CREST, enfermedad por crioaglutininas, lupus discoide, crioglobulinemia mixta esencial, fibromialgia-fibromiositis, glomerulonefritis, enfermedad de Graves, el síndrome de Guillain-Barre, tiroiditis de Hashimoto, fibrosis pulmonar idiopática, púrpura trombocitopénica idiopática (PTI), la neuropatía por IgA, artritis juvenil, el liquen plano, enfermedad de Meniere, enfermedad mixta del tejido conectivo, esclerosis múltiple , diabetes mellitus inmunomediada o tipo 1 , miastenia gravis, pénfigo vulgar, la anemia perniciosa, la poliarteritis nodosa, policondritis, síndromes poliglandulares, polimialgia reumática, la polimiositis y la dermatomiositis, agammaglobulinemia primaria, cirrosis biliar primaria, psoriasis, artritis psoriásica, el fenómeno de Raynauld , síndrome de Reiter, la sarcoidosis, la esclerodermia, la esclerosis sistémica progresiva, síndrome de Sjógren, síndrome de Goodpasture , el síndrome del hombre rígido, lupus eritematoso sistémico, lupus eritematoso, la arteritis de Takayasu, arteristis temporal/arteritis de células gigantes , colitis ulcerosa , uveítis, vasculitis como la vasculitis dermatitis herpetiforme, vitíligo , granulomatosis de Wegener, Enfermedad de la Membrana basal glomerular, síndrome antifosfolípido, Enfermedades Autoinmunes del Sistema Nervioso, fiebre mediterránea familiar, el síndrome miasténico de Lambert- Eaton, la oftalmía simpática , poliendocrinopatías , psoriasis, etc En una realización particular, dicha enfermedad autoinmune es lupus eritematoso sistémico (LES). Also part of the invention are genetic DNA constructs capable of transcribing to a peptide, antibody or antibody fragment, for use in the treatment, improvement or prevention of infectious, inflammatory or autoimmune diseases mediated by IFN. Said genetic construction of DNA would direct the in vitro or intracellular transcription of the antibody sequence or fragment thereof, and comprises at least one of the following types of sequences: a) DNA nucleotide sequence, preferably double stranded, which It comprises, at least, the coding sequence of the antibody of the invention or of the antibody fragment of the invention for in vitro, or intracellular, transcription, b) DNA nucleotide sequence, preferably double stranded, corresponding to a system or vector of gene expression comprising the sequence coding for the antibody sequence or antibody fragment of the invention operably linked with at least one promoter that directs the transcription of said nucleotide sequence of interest, and with other sequences necessary or appropriate for transcription and its proper regulation in time and place, for example, start and end signals, cut sites, polyadenylation signal, origin of replication, transcriptional activators (enhancers), transcriptional silencers (silencers), etc. for use in those pathological contexts that pass with IFN release, preferably type I and III. A "ribozyme" as understood in the present invention, refers to a catalytic polynucleotide (typically RNA), which can be constructed to specifically recognize, by hybridization, an mRNA and fragment it or eliminate its expression. Ribozymes can be introduced into the cell as catalytic RNA molecules or as genetic constructs that are expressed to RNA catalytic molecules. The term "autoimmune disease" refers to a condition in a subject characterized by a cellular, tissue and / or organ injury caused by an immunological reaction of the subject to its own cells, tissues and / or organs. Illustrative, non-limiting examples of autoimmune diseases include alopecia areata, ankylosing spondylitis, antiphospholipid syndrome, Addison autoimmune disease, autoimmune diseases of the adrenal gland, autoimmune hemolytic anemia, autoimmune hepatitis, autoimmune ooforitis and orchitis, autoimmune thrombocytopenia, autoimmune disease Behcet, bullous pemphigoid, cardiomyopathy, celiac disease, dermatitis, chronic fatigue, immune dysfunction syndrome (CFS), chronic inflammatory demyelinating polyneuropathy, Churg - Strauss syndrome, scarring penfigoid, CREST syndrome, cryogglutinin syndrome, discoid lupus , essential mixed cryoglobulinemia, fibromyalgia-fibromiositis, glomerulonephritis, Graves disease, Guillain-Barre syndrome, Hashimoto's thyroiditis, idiopathic pulmonary fibrosis, idiopathic thrombocytopenic purpura (ITP), IgA neuropathy, juvenile arthritis, lichen planus, dise Meiere de Meniere, mixed connective tissue disease, multiple sclerosis, immunomediated or type 1 diabetes mellitus, myasthenia gravis, pemphigus vulgaris, pernicious anemia, polyarteritis nodosa, polychondritis, polyglandular syndromes, polymyalgia rheumatica, polymyositis and dermatomyositis, primary agammyelitis , primary biliary cirrhosis, psoriasis, psoriatic arthritis, Raynauld's phenomenon, Reiter's syndrome, sarcoidosis, scleroderma, progressive systemic sclerosis, Sjógren's syndrome, Goodpasture's syndrome, rigid man's syndrome, lupus erythematosus, lupus erythematosus , Takayasu arteritis, temporal arteristis / giant cell arteritis, ulcerative colitis, uveitis, vasculitis such as vasculitis, herpetiform dermatitis, vitiligo, Wegener granulomatosis, Glomerular basement membrane disease, antiphospholipid syndrome, Autoimmune diseases of the Mediterranean System, fever family, the myasthenic syndrome of Lambert-Eaton, sympathetic ophthalmia, polyocrinopathies, psoriasis, etc. In a particular embodiment, said autoimmune disease is systemic lupus erythematosus (SLE).
El término "tratamiento" o "tratar" en el contexto de esta memoria descriptiva, significa la administración de un compuesto o formulación de acuerdo con la invención para prevenir, mejorar o eliminar la enfermedad o uno o más síntomas asociados con dicha enfermedad. "Tratamiento" también abarca la prevención, la mejora o la eliminación de la secuela fisiológica de la enfermedad. The term "treatment" or "treat" in the context of this specification, means the administration of a compound or formulation according to the invention to prevent, ameliorate or eliminate the disease or one or more symptoms associated with said disease. "Treatment" also covers the prevention, improvement or elimination of the physiological sequel of the disease.
El tratamiento de las enfermedades autoinmunes se hace típicamente con inmunosupresores. Se trata de sustancias que disminuyen la respuesta inmune. Ellas pueden ser ya sea exógenas, como los fármacos inmunosupresores, o endógenas. Ejemplos de fármacos inmunosupresores son los glucocorticoides, que disminuye tanto la expansión de clones de células B como la síntesis de anticuerpos; citostáticos, que afecta a la proliferación tanto de las células T como de las células B, ejemplos de agentes citostáticos sonagentes alquilantes, antimetabolitos (por ejemplo, los análogos de ácido fólico, análogos de la purina, análogos de pirimidina, inhibidores de la síntesis proteína); anticuerpos o fármacos que actúan sobre inmunofilinas como ciclosporina, tacrolimus o sirolimus. LES se trata con inmunosupresión, principalmente con ciclofosfamida o corticosteroides. The treatment of autoimmune diseases is typically done with immunosuppressants. These are substances that decrease the immune response. They can be either exogenous, such as immunosuppressive drugs, or endogenous. Examples of immunosuppressive drugs are glucocorticoids, which decreases both the expansion of B cell clones and the synthesis of antibodies; cytostatics, which affects the proliferation of both T cells and B cells, examples of alkylating agents are cytostatic agents, antimetabolites (for example, folic acid analogs, purine analogs, pyrimidine analogs, protein synthesis inhibitors ); antibodies or drugs that act on immunophilins such as cyclosporine, tacrolimus or sirolimus. SLE is treated with immunosuppression, mainly with cyclophosphamide or corticosteroids.
En otra realización preferida de este aspecto, la composición de la invención se usa para el tratamiento de una enfermedad autoinmune. En una realización más preferida de este aspecto de la invención, la enfermedad autoinmune se selecciona de la lista que consiste en: alopecia areata, la espondilitis anquilosante, síndrome antifosfolípidos, enfermedad autoinmune de Addison, enfermedades autoinmunes de la glándula suprarrenal, la anemia hemolítica autoinmune, hepatitis autoinmune, ooforitis autoinmune y orquitis, trombocitopenia autoinmune, enfermedad de Behcet, penfigoide bulloso, cardiomiopatía, enfermedad celíaca , la dermatitis , la fatiga crónica , síndrome de disfunción inmune ( SFC ), la polineuropatía desmielinizante inflamatoria crónica , síndrome de Churg -Strauss, penfigoide cicatricial, síndrome de CREST, enfermedad por crioaglutininas, lupus discoide, crioglobulinemia mixta esencial, fibromialgia- fibromiositis, glomerulonefritis, enfermedad de Graves, el síndrome de Guillain-Barre, tiroiditis de Hashimoto, fibrosis pulmonar idiopática, púrpura trombocitopénica idiopática (PTI), la neuropatía por IgA, artritis juvenil, el liquen plano, enfermedad de Meniere, enfermedad mixta del tejido conectivo, esclerosis múltiple , diabetes mellitus inmunomediada o tipo 1 , miastenia gravis, pénfigo vulgar, la anemia perniciosa, la poliarteritis nodosa, policondritis, síndromes poliglandulares, polimialgia reumática, la polimiositis y la dermatomiositis, agammaglobulinemia primaria, cirrosis biliar primaria, psoriasis, artritis psoriásica, el fenómeno de Raynauld , síndrome de Reiter, la sarcoidosis, la esclerodermia, la esclerosis sistémica progresiva, síndrome de Sjógren , síndrome de Goodpasture , el síndrome del hombre rígido, lupus eritematoso sistémico, lupus eritematoso, la arteritis de Takayasu, arteristis temporal/arteritis de células gigantes , colitis ulcerosa , uveítis, vasculitis como la vasculitis dermatitis herpetiforme, vitíligo , granulomatosis de Wegener, Enfermedad de la Membrana basal glomerular, síndrome antifosfolípido, Enfermedades Autoinmunes del Sistema Nervioso, fiebre mediterránea familiar, el síndrome miasténico de Lambert- Eaton, la oftalmía simpática , poliendocrinopatías y psoriasis. In another preferred embodiment of this aspect, the composition of the invention is used for the treatment of an autoimmune disease. In a more preferred embodiment of this aspect of the invention, the autoimmune disease is selected from the list consisting of: alopecia areata, ankylosing spondylitis, antiphospholipid syndrome, Addison autoimmune disease, autoimmune diseases of the adrenal gland, autoimmune hemolytic anemia , autoimmune hepatitis, autoimmune oophoritis and orchitis, autoimmune thrombocytopenia, Behcet's disease, bullous pemphigoid, cardiomyopathy, celiac disease, dermatitis, chronic fatigue, immune dysfunction syndrome (CFS), chronic inflammatory demyelinating polyneuropathy, Churg-Strauss syndrome , scarring pemphigoid, CREST syndrome, cryogglutinin disease, discoid lupus, essential mixed cryoglobulinemia, fibromyalgia-fibromiositis, glomerulonephritis, Graves disease, Guillain-Barre syndrome, Hashimoto's thyroiditis, idiopathic pulmonary fibrosis, idiopathic thrombocytopenic purpura , the neu IgA clothing, juvenile arthritis, lichen planus, Meniere's disease, mixed connective tissue disease, multiple sclerosis, immunomediated or type 1 diabetes mellitus, myasthenia gravis, pemphigus vulgaris, pernicious anemia, polyarteritis nodosa, polychondritis, polyglandular syndromes, polymyalgia rheumatica, polymyositis and dermatomyositis, primary agammaglobulinemia, primary biliary cirrhosis, psoriasis, psoriatic arthritis, Raynauld's phenomenon, Reiter's syndrome, sarcoidosis, scleroderma, progressive systemic sclerosis, Sjógren's syndrome, Goodpasture's syndrome, Goodpasture's syndrome rigid man syndrome, lupus systemic erythematosus, lupus erythematosus, Takayasu arteritis, temporal arteristis / giant cell arteritis, ulcerative colitis, uveitis, vasculitis such as vasculitis dermatitis herpetiformis, vitiligo, Wegener granulomatosis, Glomeripulous basal membrane disease, Autophosphine syndrome Nervous system, familial Mediterranean fever, Lambert-Eaton myasthenic syndrome, sympathetic ophthalmia, polyocrinopathies and psoriasis.
En una realización particular de la invención, la enfermedad autoinmune es el lupus eritematoso sistémico. En otra realización particular de la invención, la enfermedad autoinmune es la psoriasis. In a particular embodiment of the invention, the autoimmune disease is systemic lupus erythematosus. In another particular embodiment of the invention, the autoimmune disease is psoriasis.
La composición proporcionada por esta invención puede ser facilitada por cualquier vía de administración, para lo cual dicha composición se formulará en la forma farmacéutica adecuada a la vía de administración elegida. Los agentes moduladores de la actividad de SIDT1 de dichas composiciones se encuentran en una cantidad terapéuticamente efectiva. En el sentido utilizado en esta descripción, la expresión "cantidad terapéuticamente efectiva" se refiere a la cantidad de agentes moduladores (o construcciones genéticas que permitan su expresión intracelular) calculada para producir el efecto deseado y, en general, vendrá determinada, entre otras causas, por las características propias de dichos agentes (y construcciones) y el efecto terapéutico a conseguir. Los adyuvantes y vehículos farmacéuticamente aceptables que pueden ser utilizados en dichas composiciones son los vehículos conocidos por los técnicos en la materia. The composition provided by this invention may be provided by any route of administration, for which said composition will be formulated in the pharmaceutical form appropriate to the route of administration chosen. SIDT1 activity modulating agents of said compositions are in a therapeutically effective amount. In the sense used in this description, the term "therapeutically effective amount" refers to the amount of modulating agents (or genetic constructs that allow their intracellular expression) calculated to produce the desired effect and, in general, will be determined, among other causes. , due to the characteristics of said agents (and constructions) and the therapeutic effect to be achieved. Pharmaceutically acceptable adjuvants and vehicles that can be used in said compositions are the vehicles known to those skilled in the art.
MÉTODO DE SCREENING DE FÁRMACOS DE LA INVENCIÓN SCREENING METHOD OF DRUGS OF THE INVENTION
La invención proporciona métodos para identificar compuestos que pueden ser usados para el tratamiento de enfermedades relacionadas con SIDT1 , y en concreto enfermedades inflamatorias o autoinmunes mediadas por IFN, y preferiblemente mediadas por IFN de tipo I y III. The invention provides methods for identifying compounds that can be used for the treatment of diseases related to SIDT1, and in particular inflammatory diseases or autoimmune diseases mediated by IFN, and preferably mediated by IFN type I and III.
Estos métodos permiten la identificación de candidatos, compuestos a ensayar o agentes (por ejemplo, péptidos, peptidomiméticos, moléculas orgánicas, oligonucleótidos antisentido o otras moléculas) que pueden unirse a SIDT1 y/o tener un efecto activador o inhibidor de la actividad biológica de SIDT1 o de su expresión, y así determinar si esos compuestos tendrían un efecto sobre las enfermedades en las que SIDT1 está implicado, y en concreto aquellas enfermedades inflamatorias o autoinmunes mediadas por IFN. These methods allow the identification of candidates, compounds to be tested or agents (for example, peptides, peptidomimetics, organic molecules, antisense oligonucleotides or other molecules) that can bind SIDT1 and / or have an activating or inhibiting effect of the biological activity of SIDT1 or of its expression, and thus determine whether those compounds would have an effect on the diseases in which SIDT1 is involved, and in particular those inflammatory or autoimmune diseases mediated by IFN.
Los ensayos para identificar estas moléculas, compuestos o agentes que modulan la actividad de SIDT1 pueden emplear células que expresan SIDT1 , o en ensayos con SIDT1 aislado (o con sus variantes, como fragmentos biológicamente activos o proteínas de fusión que incluyen una porción o parte de SI DT1). Así, otro aspecto de la invención consiste en un método de selección de agentes terapéuticos útiles en el tratamiento de enfermedades inflamatorias o autoinmunes mediadas por IFN, y más específicamente para el tratamiento de la psoriasis y el lupus; que comprende: a) poner en contacto el compuesto a analizar con el polipéptido SIDT1 , y b) detectar la unión de dicho compuesto a analizar con el polipéptido SIDT1. Assays to identify these molecules, compounds or agents that modulate the activity of SIDT1 may employ cells expressing SIDT1, or in assays with isolated SIDT1 (or with its variants, such as biologically active fragments or fusion proteins that include a portion or part of YES DT1). Thus, another aspect of the invention consists in a method of selecting therapeutic agents useful in the treatment of inflammatory or autoimmune diseases mediated by IFN, and more specifically for the treatment of psoriasis and lupus; which comprises: a) contacting the compound to be analyzed with the SIDT1 polypeptide, and b) detecting the binding of said compound to be analyzed with the SIDT1 polypeptide.
Los compuestos que se unen al polipéptido SIDT1 se identificarían como agentes terapéuticos potenciales frente a enfermedades inflamatorias o autoinmunes mediadas por IFN, y más específicamente para el tratamiento de la psoriasis y el lupus. Compounds that bind to the SIDT1 polypeptide would be identified as potential therapeutic agents against inflammatory or autoimmune diseases mediated by IFN, and more specifically for the treatment of psoriasis and lupus.
Como se ha dicho, estos ensayos pueden implicar el polipéptido completo SIDT1 , un fragmento biológicamente activo del mismo, o una proteína de fusión que implique toda o una porción del polipéptido SIDT1. Determinar la capacidad de un compuesto para modular la actividad de SIDT1 puede realizarse, por ejemplo, determinando la capacidad de SIDT1 de unirse o interaccionar con una molécula diana de dicho compuesto, de manera directa o indirecta. Pueden ser también ensayos de actividad, midiendo de manera directa o indirecta la actividad de SIDT1. También puede ser un ensayo de expresión, determinando de manera directa o indirecta la expresión del mARN de SIDT1 o de la proteína SIDT1. Estos ensayos también pueden combinarse con un ensayo in vivo midiendo el efecto de un compuesto test sobre los síntomas de enfermedades relacionadas con SIDT1 , y en concreto aquellas enfermedades inflamatorias o autoinmunes mediadas por IFN, y preferiblemente mediadas por IFN de tipo I y III (por ejemplo, pero sin limitarse, sobre modelos animales u otros sistemas modelo conocidos en la técnica). As stated, these assays may involve the complete SIDT1 polypeptide, a biologically active fragment thereof, or a fusion protein that involves all or a portion of the SIDT1 polypeptide. Determining the ability of a compound to modulate the activity of SIDT1 can be performed, for example, by determining the ability of SIDT1 to bind or interact with a target molecule of said compound, directly or indirectly. They can also be activity tests, directly or indirectly measuring the activity of SIDT1. It can also be an expression assay, directly or indirectly determining the expression of the SIDT1 mRNA or SIDT1 protein. These tests can also be combined with an in vivo test by measuring the effect of a test compound on the symptoms of SIDT1-related diseases, and in particular those inflammatory or autoimmune diseases mediated by IFN, and preferably mediated by IFN type I and III (by example, but not limited, on animal models or other model systems known in the art).
Los compuestos a testar empleados en el método de selección de agentes terapéuticos no se limitan a moléculas orgánicas de bajo peso molecular, proteínas (incluyendo anticuerpos), péptidos, oliogonucleótidos, etc. Pueden ser naturales y/o sintéticos. Por ejemplo, anticuerpos capaces de unirse a un epítopo de SIDT1 , que pueden ser empleados terapéuticamente, como se ha expuesto anteriormente, pueden emplearse también en ensayos inmunohistoquímicos, como Western blots, ELISAs, radioinmunoensayos, ensayos de inmunoprecipitación, o otros ensayos inmunohistoquímicos conocidos en el estado de la técnica. SIDT1 puede emplearse para inmunizar a un animal, para obtener anticuerpos policlonales. También se pueden preparar anticuerpos monoclonales mediante técnicas que permiten la producción de anticuerpos por líneas celulares en cultivo, entre las que se incluyen, pero sin limitarse, hibridomas, hibridomas de células B humanas o colecciones de nanobodies. Técnicas para producir anticuerpos quiméricos, humanizados o sintéticos son conocidas. Los agentes terapéuticos identificados por el método de selección aquí descrito pueden ser usados en un modelo animal o de otro tipo para determinar el mecanismo de acción de dicho agente. Más aún, los agentes terapéuticos seleccionados por el método aquí descrito se emplearían en el tratamiento de enfermedades que cursen con la alteración de SIDT1 y, en concreto, enfermedades inflamatorias o autoinmunes mediadas por IFN, preferiblemente por IFN de tipo I y III, y más específicamente la psoriasis y el lupus. The compounds to be tested used in the method of selection of therapeutic agents are not limited to low molecular weight organic molecules, proteins (including antibodies), peptides, oliogonucleotides, etc. They can be natural and / or synthetic. For example, antibodies capable of binding to an epitope of SIDT1, which can be used therapeutically, as discussed above, can also be used in immunohistochemical assays, such as Western blots, ELISAs, radioimmunoassays, immunoprecipitation assays, or other immunohistochemical assays known in The state of the art. SIDT1 can be used to immunize an animal, to obtain polyclonal antibodies. Monoclonal antibodies can also be prepared by techniques that allow the production of antibodies by cultured cell lines, including, but not limited to, hybridomas, human B-cell hybridomas or collections of nanobodies. Techniques for producing chimeric, humanized or synthetic antibodies are known. The therapeutic agents identified by the selection method described herein can be used in an animal or other model to determine the mechanism of action of said agent. Moreover, the therapeutic agents selected by the method described herein would be used in the treatment of diseases that occur with the alteration of SIDT1 and, in particular, IFN-mediated inflammatory or autoimmune diseases, preferably by type I and III IFNs, and more specifically psoriasis and lupus.
En otro aspecto de la invención se describe un método de selección de agentes terapéuticos útiles en el tratamiento de enfermedades inflamatorias o autoinmunes mediadas por IFN, preferiblemente por IFN de tipo I y III, y más específicamente para el tratamiento de la psoriasis y el lupus, que comprende: a) determinar la actividad de SIDT1 a una concentración establecida del compuesto a analizar o en ausencia de dicho compuesto, b) determinar la actividad de SIDT1 a una concentración del compuesto a analizar diferente de la de a). Compuestos que den lugar a una actividad diferente de SIDT1 se identificarían como agentes terapéuticos potenciales frente a enfermedades inflamatorias o autoinmunes mediadas por IFN, preferiblemente por IFN de tipo I y III, y más específicamente para la psoriasis y el lupus. In another aspect of the invention a method of selection of therapeutic agents useful in the treatment of inflammatory diseases or autoimmune diseases mediated by IFN, preferably by type I and III IFN, and more specifically for the treatment of psoriasis and lupus, is described. which comprises: a) determining the activity of SIDT1 at an established concentration of the compound to be analyzed or in the absence of said compound, b) determining the activity of SIDT1 at a concentration of the compound to be analyzed different from that of a). Compounds that give rise to a different activity of SIDT1 would be identified as potential therapeutic agents against inflammatory or autoimmune diseases mediated by IFN, preferably by type I and III IFN, and more specifically for psoriasis and lupus.
Otro aspecto de la invención consiste en un método de selección de agentes terapéuticos útiles en el tratamiento de enfermedades inflamatorias o autoinmunes mediadas por IFN, preferiblemente por IFN de tipo I y III, y más específicamente de la psoriasis y el lupus, que comprende: a) poner en contacto el compuesto a analizar con el polinucleótido SIDT1 , b) detectar la unión de dicho compuesto a analizar con el polinucleótido SIDT1 Another aspect of the invention consists in a method of selecting therapeutic agents useful in the treatment of inflammatory or autoimmune diseases mediated by IFN, preferably by type I and III IFN, and more specifically psoriasis and lupus, comprising: a ) contacting the compound to be analyzed with the SIDT1 polynucleotide, b) detecting the binding of said compound to be analyzed with the SIDT1 polynucleotide
Los compuestos que se unen al polinucleótido SIDT1 se identificarían como agentes terapéuticos potenciales frente a enfermedades inflamatorias o autoinmunes mediadas por IFN, preferiblemente por IFN de tipo I y III, y más específicamente de la psoriasis y el lupus. Compounds that bind to the SIDT1 polynucleotide would be identified as potential therapeutic agents against inflammatory or autoimmune diseases mediated by IFN, preferably by type I and III IFN, and more specifically from psoriasis and lupus.
MÉTODO DE DIAGNÓSTICO DE LA INVENCIÓN METHOD OF DIAGNOSIS OF THE INVENTION
Las enfermedades en las que la alteración de la actividad de SIDT1 puede ser diagnóstica, y en concreto enfermedades inflamatorias o autoinmunes mediadas por IFN, y más específicamente la psoriasis y el lupus, pueden ser detectadas midiendo la cantidad de ácidos nucleicos (ADN y/o ARN y/o mARN) que codifican para SIDT1 , o la cantidad de proteína SIDT1 que se expresa, en comparación con células normales. La detección de los oligonucleótidos puede hacerse por métodos bien conocidos en el estado de la técnica (como por ejemplo, pero sin limitarse, sondas con nucleótidos marcados, hibridación ADN-ADN ó ADN-ARN, amplificación por PCR empleando nucleótidos marcados, la RT-PCR). Procedimientos para detectar la expresión de la proteína SIDT1 también son bien conocidos en el estado de la técnica, como por ejemplo, pero sin limitarse, anticuerpos poli o monoclonales, ELISA, radioinmunoensayo (RIA), y FACS (fluorescence activated cell sorting). Diseases in which the alteration of the activity of SIDT1 can be diagnostic, and in particular inflammatory diseases or autoimmune mediated by IFN, and more specifically psoriasis and lupus, can be detected by measuring the amount of nucleic acids (DNA and / or RNA and / or mRNA) encoding SIDT1, or the amount of SIDT1 protein that is expressed, compared to normal cells. Oligonucleotide detection it can be done by methods well known in the state of the art (such as, but not limited to, probes with labeled nucleotides, DNA-DNA or DNA-RNA hybridization, PCR amplification using labeled nucleotides, RT-PCR). Methods for detecting the expression of the SIDT1 protein are also well known in the state of the art, such as, but not limited to, poly or monoclonal antibodies, ELISA, radioimmunoassay (RIA), and FACS (fluorescence activated cell sorting).
Por tanto, en otro aspecto de la invención se describe un método para la recolección de datos útiles en el diagnóstico y/o pronóstico de enfermedades inflamatorias o autoinmunes mediadas por IFN, y más específicamente de la psoriasis y el lupus, que comprende: a) determinar la expresión de SIDT1 en una muestra extraída de un mamífero, b) comparar los valores de la expresión de SIDT1 obtenidos en a) con los valores estándar en mamíferos sanos o enfermos. Therefore, in another aspect of the invention a method is described for the collection of data useful in the diagnosis and / or prognosis of inflammatory or autoimmune diseases mediated by IFN, and more specifically psoriasis and lupus, comprising: a) determine the expression of SIDT1 in a sample taken from a mammal, b) compare the values of the expression of SIDT1 obtained in a) with the standard values in healthy or diseased mammals.
Entre las enfermedades inflamatorias o autoinmunes mediadas por IFN se incluyen preferiblemente, pero sin limitarse, el lupus eritematoso sistémico, esclerosis sistémica, psoriasis, síndrome de Sjógren, artritis reumatoide, síndrome de Accardi-Goutiéres, alopecia areata, la espondilitis anquilosante, síndrome antifosfolípidos, enfermedad autoinmune de Addison, enfermedades autoinmunes de la glándula suprarrenal, la anemia hemolítica autoinmune, hepatitis autoinmune, ooforitis autoinmune y orquitis, trombocitopenia autoinmune, enfermedad de Behcet, penfigoide bulloso, cardiomiopatía, enfermedad celíaca , la dermatitis , la fatiga crónica , síndrome de disfunción inmune ( SFC ), la polineuropatía desmielinizante inflamatoria crónica , síndrome de Churg -Strauss, penfigoide cicatricial, síndrome de CREST, enfermedad por crioaglutininas, lupus discoide, crioglobulinemia mixta esencial, fibromialgia- fibromiositis, glomerulonefritis, enfermedad de Graves, el síndrome de Guillain-Barre, tiroiditis de Hashimoto, fibrosis pulmonar idiopática, púrpura trombocitopénica idiopática (PTI), la neuropatía por IgA, artritis juvenil, el liquen plano, enfermedad de Meniere, enfermedad mixta del tejido conectivo, esclerosis múltiple , diabetes mellitus inmunomediada o tipo 1 , miastenia gravis, pénfigo vulgar, la anemia perniciosa, la poliarteritis nodosa, policondritis, síndromes poliglandulares, polimialgia reumática, la polimiositis y la dermatomiositis, agammaglobulinemia primaria, cirrosis biliar primaria, psoriasis, artritis psoriásica, el fenómeno de Raynauld, síndrome de Reiter, la sarcoidosis, la esclerodermia, la esclerosis sistémica progresiva, síndrome de Goodpasture, el síndrome del hombre rígido, lupus eritematoso sistémico, lupus eritematoso, la arteritis de Takayasu, arteristis temporal/arteritis de células gigantes , colitis ulcerosa , uveítis, vasculitis como la vasculitis dermatitis herpetiforme, vitíligo , granulomatosis de Wegener, Enfermedad de la Membrana basal glomerular, síndrome antifosfolípido, Enfermedades Autoinmunes del Sistema Nervioso, fiebre mediterránea familiar, el síndrome miasténico de Lambert- Eaton, la oftalmía simpática y poliendocrinopatías. Los términos "polinucleótido" y "ácido nucleico" se usan aquí de manera intercambiable, refiriéndose a formas poliméricas de nucleótidos de cualquier longitud, tanto ribonucleótidos como desoxiribonucleótidos. IFN-mediated inflammatory or autoimmune diseases include, but are not limited to, systemic lupus erythematosus, systemic sclerosis, psoriasis, Sjógren syndrome, rheumatoid arthritis, Accardi-Goutiéres syndrome, alopecia areata, ankylosing spondylitis, antiphospholipid syndrome, antiphospholipid syndrome Addison autoimmune disease, autoimmune diseases of the adrenal gland, autoimmune hemolytic anemia, autoimmune hepatitis, autoimmune oophoritis and orchitis, autoimmune thrombocytopenia, Behcet's disease, bullous pemphigoid, cardiomyopathy, celiac disease, dermatitis, chronic fatigue, dysfunction syndrome Immune (CFS), chronic inflammatory demyelinating polyneuropathy, Churg-Strauss syndrome, scarring penfigoid, CREST syndrome, cryoagglutinin disease, discoid lupus, essential mixed cryoglobulinemia, fibromyalgia-fibromiositis, glomerulonephritis, Graves disease, syndrome Guillain-Barre, Hashimoto's thyroiditis, idiopathic pulmonary fibrosis, idiopathic thrombocytopenic purpura (ITP), IgA neuropathy, juvenile arthritis, lichen planus, Meniere's disease, mixed connective tissue disease, multiple sclerosis, immunomediated diabetes mellitus or type 1 Myasthenia gravis, pemphigus vulgaris, pernicious anemia, polyarteritis nodosa, polychondritis, polyglandular syndromes, polymyalgia rheumatica, polymyositis and dermatomyositis, primary agammaglobulinemia, primary biliary cirrhosis, psoriasis, psoriatic arthritis, Raynaiter's phenomenon, Raynaiter's phenomenon sarcoidosis, scleroderma, progressive systemic sclerosis, Goodpasture syndrome, rigid man syndrome, systemic lupus erythematosus, lupus erythematosus, Takayasu arteritis, temporal arteristis / giant cell arteritis, ulcerative colitis, uveitis, vasculitis such as vasculitis dermatitis herpetiformis, vitiligo, W granulomatosis egener, Glomerular basement membrane disease, antiphospholipid syndrome, Autoimmune Diseases of the Nervous System, family Mediterranean fever, Lambert-Eaton myasthenic syndrome, sympathetic ophthalmia and polyocrinopathies. The terms "polynucleotide" and "nucleic acid" are used interchangeably herein, referring to polymeric forms of nucleotides of any length, both ribonucleotides and deoxyribonucleotides.
Los términos "péptido", "oligopéptido", "polipéptido" y "proteína" se usan aquí de manera intercambiable, y se refieren a una forma polimérica de aminoácidos de cualquier longitud, que pueden ser codificantes o no codificantes, química o bioquímicamente modificados. The terms "peptide", "oligopeptide", "polypeptide" and "protein" are used interchangeably herein, and refer to a polymeric form of amino acids of any length, which may be coding or non-coding, chemically or biochemically modified.
A lo largo de la descripción y las reivindicaciones la palabra "comprende" y sus variantes no pretenden excluir otras características técnicas, aditivos, componentes o pasos. Para los expertos en la materia, otros objetos, ventajas y características de la invención se desprenderán en parte de la descripción y en parte de la práctica de la invención. Los siguientes ejemplos y dibujos se proporcionan a modo de ilustración, y no se pretende que sean limitativos de la presente invención. Throughout the description and the claims the word "comprises" and its variants are not intended to exclude other technical characteristics, additives, components or steps. For those skilled in the art, other objects, advantages and features of the invention will be derived partly from the description and partly from the practice of the invention. The following examples and drawings are provided by way of illustration, and are not intended to be limiting of the present invention.
DESCRIPCIÓN DE LAS FIGURAS DESCRIPTION OF THE FIGURES
Fig. 1. Expresión del gen SIDT1 en varias líneas celulares. A) Medida de la abundancia del mARN SIDT1 en distintas líneas celulares mediante qRT-PCR: Hela y JEK-293, células epiteliales; K562: serie granulocítica/pre-eritrocítica; THP-1 : monocitos; HL-60: promieloblasto; daudi, ramos, RL, raji, namalwa: linfocito B; Jurkat: linfocito T; NKL: células NK; Gen2.2: pDC. B) Sobreexpresión de SIDT1 en la línea celular Gen2.2 tras estimulación con ligandos de TLR9 (ODN2006) y TKR7 (imiquimod) medida mediante qRT-PCR. Se indica la expresión del gen SIDT1 usando como referencia la expresión del gen HPRT1. Fig. 1. Expression of the SIDT1 gene in several cell lines. A) Measurement of the abundance of the SIDT1 mRNA in different cell lines using qRT-PCR: Hela and JEK-293, epithelial cells; K562: granulocytic / pre-erythrocytic series; THP-1: monocytes; HL-60: promyloblast; daudi, bouquets, RL, raji, namalwa: B lymphocyte; Jurkat: T lymphocyte; NKL: NK cells; Gen2.2: pDC. B) Overexpression of SIDT1 in the Gen2.2 cell line after stimulation with TLR9 (ODN2006) and TKR7 (imiquimod) ligands measured by qRT-PCR. The expression of the SIDT1 gene is indicated using as reference the expression of the HPRT1 gene.
Fig. 2. Genes regulados negativamente en las células Gen2.2 silenciadas para SIDT1 , tras estimulación con ODN2006 durante 6h. La expression de los diferentes genes se realizó mediante arrays de expresión. En la tabla se expresa la expresión relativa en las células silenciadas frente a las células tratadas con un shRNA control, sin diana. En la tabla se representan los 50 primeros genes ordenados por importancia de efecto. Fig. 2. Negatively regulated genes in Gen2.2 cells silenced for SIDT1, after stimulation with ODN2006 for 6h. The expression of the different genes was carried out by means of expression arrays. The table shows the relative expression in the silenced cells versus the cells treated with a control shRNA, without target. The table shows the first 50 genes sorted by importance of effect.
Fig. 3. Producción de citoquinas pro-inflamatorias en células Gen2.2 shSIDTI tras estimulación con ligandos de TLR) (ODN2006) y TLR7 (Imiquimod). Las células se estimularon durante 24h con ODN2006 o imiquimod y se midió la cantidad de TNFa, IL-6 e IL-8 en el sobrenadante mediante ELISA. Como control se incluyeron células no trasducidas, y células trasducidas con un shRNA control. Fig. 4. Producción de moléculas de la vía del IFN en células gen2.2 silenciadas para SIDT1 estimuladas. Las células gen2.2 shSIDTI y sus controles se estimularon como en la figura 3 y la producción interferones de tipo I (IFNal) y III (IFNI1) y de la quimioquina dependiente de IFN CCL3 se midió mediante ELISA. Fig. 5. Inhibición parcial de la entrada de oligonucleótidos en la célula mediante silenciamiento de SIDT1. Las células gen2.2 shSIDTI se incubaron con el oligonucleótido estimulador de TLR9 marcado ODN2006-Cy5 y se midió su entrada en la célula mediante citometría de flujo. Como control se incluyeron células no trasducidas, y células trasducidas con un shRNA control. Fig. 6. El silenciamiento de SIDT1 bloquea la producción de IFN de tipo I por pDC aisladas ex vivo en respuesta a ODN2006. A) Medida de la expresión del gen SIDT1 en pDC circulantes trasducidas con vectores lentivirales shSIDTI Las pDC se aislaron de sangre de donantes sanos mediante métodos inmunomagnéticos. La expresión se midió mediante técnicas de branch-DNA, utilizando como RNA control HMBS. B) Producción de IFN de tipo I en las pDC silenciadas tras estimulación con ODN2006 ex vivo. La concentración se determinó mediante el uso de células indicadoras tras 24h de estimulación. Fig. 3. Production of pro-inflammatory cytokines in Gen2.2 shSIDTI cells after stimulation with TLR ligands) (ODN2006) and TLR7 (Imiquimod). The cells were stimulated for 24h with ODN2006 or imiquimod and the amount of TNFa, IL-6 and IL-8 in the supernatant was measured by ELISA. As a control, non-transduced cells and transduced cells with a control shRNA were included. Fig. 4. Production of IFN pathway molecules in silenced gen2.2 cells for stimulated SIDT1. The gen2.2 shSIDTI cells and their controls were stimulated as in Figure 3 and the production of interferons of type I (IFNal) and III (IFNI1) and of the IFN-dependent chemokine CCL3 was measured by ELISA. Fig. 5. Partial inhibition of oligonucleotide entry into the cell by silencing SIDT1. The gen2.2 shSIDTI cells were incubated with the TLR9 stimulating oligonucleotide labeled ODN2006-Cy5 and their entry into the cell was measured by flow cytometry. As a control, non-transduced cells and transduced cells with a control shRNA were included. Fig. 6. SIDT1 silencing blocks the production of type I IFN by pDC isolated ex vivo in response to ODN2006. A) Measurement of SIDT1 gene expression in circulating pDCs transduced with shSIDTI lentiviral vectors The pDCs were isolated from blood from healthy donors by immunomagnetic methods. Expression was measured by branch-DNA techniques, using as HMBS control RNA. B) Production of type I IFN in the silenced pDCs after stimulation with ODN2006 ex vivo. The concentration was determined by the use of indicator cells after 24h of stimulation.
Fig. 7: Efecto del silenciamiento de SIDT1 en la secreción de IFN de tipo I tras estimulación de receptores endosomales y citosólicos de DNA y RNA. Las células Gen2.2 shSIDTI y sus controles se estimularon con ODN2006 o poly l:C en presencia o no de lipofectamina. La actividad de IFN de tipo I se midió en el sobrenadante tras 24h de estimulación mediante el uso de células indicadoras. Fig. 7: Effect of SIDT1 silencing on type I IFN secretion after stimulation of DNA and RNA endosomal and cytosolic receptors. Gen2.2 shSIDTI cells and their controls were stimulated with ODN2006 or poly l: C in the presence or absence of lipofectamine. Type I IFN activity was measured in the supernatant after 24h of stimulation by the use of indicator cells.
Fig. 8: Fosforilación de TBK-1 y elF2a tras estimulación de TLR9. Las células Gen2.2 shSIDTI y sus controles se estimularon con ODN2006 y se midió la fosforilación de TBK-1 y elF2a mediante Western blot. La proteína actina se utilizó para estimar la cantidad de proteínas totales en cada uno de los carriles. Fig. 8: Phosphorylation of TBK-1 and F2a after stimulation of TLR9. Gen2.2 shSIDTI cells and their controls were stimulated with ODN2006 and phosphorylation of TBK-1 and elF2a was measured by Western blot. Actin protein was used to estimate the amount of total proteins in each of the lanes.
EXPOSICIÓN DETALLADA DE MODOS DE REALIZACIÓN DETAILED EXHIBITION OF REALIZATION MODES
Los autores de la presente invención han comprobado que el mRNA de SIDT1 se expresa en líneas celulares hematopoyéticas de tipo linfoide (Fig. lA): linfocitos T y B, células NK y células dendríticas plasmacitoides (pDC). Su estudio se centra en las pDC, que son células claves para la organización de la respuesta inmune, debido a que son células presentadoras del antígeno con una alta capacidad de producción de citoquinas en respuesta a estímulos microbianos. Las pDC detectan los agentes infecciosos o las células muertas a través de receptores endosomales de ácidos nucleicos exógenos, que reconocen secuencias de RNA (Toll-like receptor 7, TLR7), o DNA con motivos CpG no metilados (TLR9). Las pDC son conocidas como las células naturales productoras de interferón (IFN), debido a su elevadísima capacidad para producir grandes cantidades de IFN de tipo I (a y β) y III (ΙΡΝλ), citoquinas fundamentales para el controlar la infección, así como para modular la actividad de los linfocitos T y B. Dada la baja frecuencia de pDC en circulación y a las dificultades para su cultivo, la mayoría de resultados obtenidos en nuestro estudio se han obtenido utilizando la línea celular Gen2.2, obtenida de un paciente con un neoplasma con fenotipo pDC. Esta línea celular constituye actualmente el principal modelo celular para el estudio de la biología de las pDC. Según los resultados, el mRNA de SIDT1 se induce tras estimulación con ligandos de TLR7 (imiquimod) y TLR9 (ODN2006) (Figura 1 B) sugiriendo un papel de SIDT1 en la respuesta mediada por estos receptores. The authors of the present invention have verified that the SIDT1 mRNA is expressed in lymphoid-type hematopoietic cell lines (Fig. 1A): T and B lymphocytes, NK cells and plasmacytoid dendritic cells (pDC). His study focuses on pDCs, which are key cells for the organization of the immune response, because they are antigen presenting cells with a high capacity for cytokine production in response to microbial stimuli. The pDCs detect infectious agents or dead cells through exogenous nucleic acid endosomal receptors, which recognize RNA sequences (Toll-like receptor 7, TLR7), or DNA with non-methylated CpG motifs (TLR9). The pDCs are known as the natural interferon-producing cells (IFN), due to their very high capacity to produce large amounts of type I (a and β) and III (ΙΡΝλ) IFN, fundamental cytokines for controlling infection, as well as for Modulate the activity of T and B lymphocytes. Given the low frequency of circulating pDC and the difficulties for its cultivation, most of the results obtained in our study have been obtained using the Gen2.2 cell line, obtained from a patient with a Neoplasm with pDC phenotype. This cell line is currently the main cell model for the study of the biology of the pDC. According to the results, the SIDT1 mRNA is induced after stimulation with TLR7 (imiquimod) and TLR9 (ODN2006) ligands (Figure 1B) suggesting a role of SIDT1 in the response mediated by these receptors.
Se realizaron arrays de expresión utilizando células Gen2.2 transducidas con vectores lentivirales portando secuencias shRNA específicas para SIDT1 , utilizando como control shRNA sin diana. Como se puede apreciar en la figura 2, los genes reprimidos en las células shSIDTI tras estimulación con ODN2006 están enriquecidos en genes relacionados con el sistema inmune, concretamente con la vía del IFN. Estos resultados indican que SIDT1 jugaría un papel central en la regulación de la vía del interferón en respuesta a la estimulación de TLR9. Expression arrays were performed using Gen2.2 cells transduced with lentiviral vectors carrying shRNA sequences specific for SIDT1, using shRNA control without target. As can be seen in Figure 2, the repressed genes in shSIDTI cells after stimulation with ODN2006 are enriched in genes related to the immune system, specifically with the IFN pathway. These results indicate that SIDT1 would play a central role in the regulation of the interferon pathway in response to stimulation of TLR9.
Para verificar ese punto, se estudió la producción de citoquinas en células Gen2.2 silenciadas y sus controles, tras estimulación con ligandos de TLR7 (imiquimod) y TLR9 (ODN2006), encontrándose que la secreción de citoquinas proinflamatorias (TNFa, IL-6, IL-8), no se encuentra afectada (Fig. 3), pero sí fuertemente la producción de IFN de tipo I (IFNal) y III (IFN l), y de CCL3, una quimioquina dependiente de IFN (Fig. 4). Mediante estudios de citometría de flujo se pudo cuantificar la entrada del oligonucleótido ODN2006 acoplado al marcador fluorescente Cy5 (Fig. 5), y pudo observarse una inhibición parcial de la internalización del ligando fluorescente, lo que no es suficiente para justificar el marcado efecto sobre la secreción de IFN. Finalmente, se realizaron experimentos sobre pDC primarias circulantes, obtenidas ex vivo mediante métodos inmunomagnéticos a partir de muestras de sangre de donantes sanos. Como se muestra en la figura 6, los resultados obtenidos con la línea celular modelo se confirman usando células primarias, ya que en las pDC silenciadas con los vectores lentivirales shSIDTI (Fig. 6A) y estimuladas con ODN2006, se observó una inhibición en la producción de IFN de tipo I de tres logaritmos de magnitud (Fig. 6B). To verify this point, the production of cytokines in silenced Gen2.2 cells and their controls was studied, after stimulation with TLR7 (imiquimod) and TLR9 (ODN2006) ligands, finding that proinflammatory cytokine secretion (TNFa, IL-6, IL-8), is not affected (Fig. 3), but strongly the production of type I IFN (IFNal) and III (IFN 1), and CCL3, an IFN-dependent chemokine (Fig. 4). Through flow cytometry studies, the input of the ODN2006 oligonucleotide coupled to the Cy5 fluorescent marker could be quantified (Fig. 5), and a partial inhibition of the internalization of the fluorescent ligand could be observed, which is not sufficient to justify the marked effect on the IFN secretion. Finally, experiments were carried out on circulating primary pDC, obtained ex vivo by immunomagnetic methods from blood samples from healthy donors. As shown in Figure 6, the results obtained with the model cell line are confirmed using primary cells, since in pDCs silenced with shSIDTI lentiviral vectors (Fig. 6A) and stimulated with ODN2006, production inhibition was observed. of type I IFN of three logarithms of magnitude (Fig. 6B).
Ejemplo de la invención Example of the invention
Materiales y métodos Expresión de SIDT1 en líneas celulares. Las líneas celulares HeLa, HEK 293T, K562, THP-1 , HL-60, daudi, RL, Raji, Namalwa, Jurkat, NKL, Gen2.2 se cultivaron en medio RMPI suplementado con 10% de suero fetal bovino a 37 °C y 7% de C02. Estas líneas celulares se utilizaron para la purificación de RNA total utilizando el kit High Puré RNA (Roche). 1 μg de RNA se utilizó como molde para la síntesis de cDNA usando el kit de Roche Transcriptor First- Strand cDNA, y a continuación se realizaron experimentos de PCR cuantitativa (qPCR) utilizando sondas taqman específicas de SIDT1 (Hs00214475_m1) y HPRT1 (Hs01003268_g1). Como se ilustra en la figura 1A, SIDT1 se expresa de forma preferencial en líneas celulares del lineaje linfoide: linfocitos T y B, células NK y células dendríticas plasmacitoides (pDC). Dado el papel central de regulación del sistema inmune de las pDC, se eligió este tipo celular como modelo para estudiar la función de SIDT1. Por ello se estimularon las células Gen2.2 con ligandos de los sensores endosomales de ácidos nucleicos TLR-7 (imiquimod 3 μg/ml) y TLR-9 (ODN2006, 1 mM) durante 24 y 48h. Los resultados de qPCR de la figura 1 B muestran una fuerte sobreexpresión de SIDT1 tras estimulación, indicando un papel de SIDT1 en la respuesta inmune mediada por estos receptores. Materials and methods Expression of SIDT1 in cell lines. The HeLa, HEK 293T, K562, THP-1, HL-60, daudi, RL, Raji, Namalwa, Jurkat, NKL, Gen2.2 cell lines were grown in RMPI medium supplemented with 10% fetal bovine serum at 37 ° C and 7% of C0 2 . These cell lines were used for purification of total RNA using the High Puré RNA kit (Roche). 1 μg of RNA was used as a template for cDNA synthesis using the Roche Transcriptor First-Strand cDNA kit, and then quantitative PCR experiments (qPCR) were performed using taqman probes specific to SIDT1 (Hs00214475_m1) and HPRT1 (Hs01003268_g1). As illustrated in Figure 1A, SIDT1 is preferentially expressed in lymphoid line cell lines: T and B lymphocytes, NK cells and plasmacytoid dendritic cells (pDC). Given the central role of regulating the immune system of the pDCs, this cell type was chosen as a model to study the function of SIDT1. Therefore, Gen2.2 cells were stimulated with ligands of the endosomal sensors of TLR-7 (imiquimod 3 μg / ml) and TLR-9 nucleic acids (ODN2006, 1 mM) for 24 and 48h. The results of qPCR in Figure 1 B show a strong overexpression of SIDT1 after stimulation, indicating a role of SIDT1 in the immune response mediated by these receptors.
Expresión diferencial de genes en células Gen2.2 silenciadas para SIDT1. Differential expression of genes in silenced Gen2.2 cells for SIDT1.
Con el objeto de estudiar el papel de SIDT1 en la respuesta mediada por TLR-7 y -9 en las pDC, se realizaron trasducciones con vectores lentivirales portando tres secuencias de shRNA específicas para SIDT1 (shRNA#1 AGCCGTGAGGGTGTATGTGAA, (SEQ ID NO: 3); shRNA#2 CTACTTGGGATGCCTTCTTGT (SEQ ID NO: 4); shRNA#3 GTGCATTCTGCTGGATTTCTT (SEQ ID NO: 5)), clonados en el vector pLenti-H1 (AMSBIO). Como control se utilizó un lentivirus portando una secuencia shRNA sin diana (shCTL GTCTCCACGCGCAGTACATTT (SEQ ID NO: 6)). En todos los casos se utilizó una multiplicidad de infección (moi) de 1. Tanto las células Gen2.2 como las células trasducidas con los tres shRNA específicos para SIDT1 (shSIDTI) como su shRNA control (shCTL) se estimularon durante 6h con 1 μΜ ODN2006. Se purificó RNA tras la estimulación y se utilizó para estudios de arrays de expresión de RNA. In order to study the role of SIDT1 in the response mediated by TLR-7 and -9 in the pDC, transductions were performed with lentiviral vectors carrying three shRNA sequences specific for SIDT1 (shRNA # 1 AGCCGTGAGGGTGTATGTGAA, (SEQ ID NO: 3 ); shRNA # 2 CTACTTGGGATGCCTTCTTGT (SEQ ID NO: 4); shRNA # 3 GTGCATTCTGCTGGATTTCTT (SEQ ID NO: 5)), cloned into the vector pLenti-H1 (AMSBIO). As a control, a lentivirus was used carrying a shRNA sequence without a target (shCTL GTCTCCACGCGCAGTACATTT (SEQ ID NO: 6)). In all cases a multiplicity of infection (moi) of 1 was used. Both Gen2.2 cells and cells transduced with The three shRNAs specific for SIDT1 (shSIDTI) as their control shRNA (shCTL) were stimulated for 6h with 1 μΜ ODN2006. RNA was purified after stimulation and was used for studies of RNA expression arrays.
El mareaje de la muestra se realizó mediante el kit de Ambion, lllumina ® RNA TotalPrep (Life Technologies), para obtener un cRNA marcado con biotina. Este cRNA se híbrido a la matriz del array de expresión de lllumina HumanHT 12v4 de transcriptoma completo, y posteriormente se reveló utilizando estreptavidina-Cy3. Sample mapping was performed using the Ambion kit, lllumina ® TotalPrep RNA (Life Technologies), to obtain a biotin-labeled cRNA. This cRNA hybridized to the array of the human transcriptome lllumine expression array 12v4, and was subsequently revealed using streptavidin-Cy3.
Los datos crudos fueron generados usando el software GenomeStudio (lllumina) y analizados posteriormente en el entorno estadístico R y Bioconductor. Los valores de expresión de cada sonda fueron determinados tras la corrección de la señal de fondo usando los controles negativos y posteriormente se aplicó una normalización por cuantiles para eliminar variaciones técnicas. Las sondas con valores de detección p<0.01 en al menos el 10% de las muestras fueron eliminados del análisis y se computó el valor medio de la expresión de cada gen aplicando la mediana al valor de todas las sondas que codificaban el mismo gen. El análisis de expresión diferencial se llevo a cabo mediante el uso del paquete Limma. Los genes que mostraron un p-valor corregido menor que 0.05 fueron considerados estadísticamente significativos. En la figura 2 se presentan los 50 genes más reprimidos en las células Gen2.2 shSIDTI estimuladas en relación con las shCTL. The raw data were generated using the GenomeStudio software (lllumina) and subsequently analyzed in the R and Bioconductor statistical environment. The expression values of each probe were determined after correction of the background signal using the negative controls and subsequently quantile normalization was applied to eliminate technical variations. Probes with detection values p <0.01 in at least 10% of the samples were removed from the analysis and the average value of the expression of each gene was computed by applying the median to the value of all probes encoding the same gene. Differential expression analysis was carried out using the Limma package. Genes that showed a corrected p-value less than 0.05 were considered statistically significant. Figure 50 shows the 50 most repressed genes in Gen2.2 shSIDTI cells stimulated in relation to shCTL.
El análisis funcional de los genes expresados diferencialmente se realizó utilizando la herramienta Genecodis. Las anotaciones con p-valores corregidos menores de 1 10"4 fueron consideradas estadísticamente enriquecidas en las listas de genes inhibidos y usadas para entender los procesos biológicos subyacentes al modelo experimental. La vías más representadas corresponden a la llamada "firma de interferón", caracterizada por genes conectados con la producción de interferones de tipo I (IFNa/β) (Rónblom y Eloranta, Clin Opin Rheumatol). Estos datos indican el papel de SIDT1 en la inducción de genes relacionados con IFN en respuesta a la estimulación de TLR-7 y -9. Functional analysis of differentially expressed genes was performed using the Genecodis tool. The annotations with corrected p-values less than 1 10 "4 were considered statistically enriched in the lists of inhibited genes and used to understand the biological processes underlying the experimental model. The most represented pathways correspond to the so-called" interferon signature ", characterized by genes connected with the production of type I interferons (IFNa / β) (Rónblom and Eloranta, Clin Opin Rheumatol) These data indicate the role of SIDT1 in the induction of IFN-related genes in response to stimulation of TLR-7 and -9.
Respuesta a la estimulación de TLR-7 y -9 en Gen2.2 shSIDTI. Response to stimulation of TLR-7 and -9 in Gen2.2 shSIDTI.
Para corroborar el resultado del array de expresión, se realizaron estimulaciones con ODN2006 y imiquimod sobre las células Gen2.2 y sus controles durante 24h, y se midieron varias citoquinas y quimioquinas en el sobrenadante mediante ensayos de ELISA (eBioscience), siguiendo el protocolo recomendado por el fabricante. Como se ilustra en la figura 3, la estimulación de la producción de las citoquinas proinflamatorias TNFa, IL-6 e IL-8 no se altera por el silenciamiento de SIDT1. Sin embargo, se observa una drástica disminución de interferón de tipo I (IFNal) y III (IFN 1), así como de la quimioquina CCL3, que está regulada por IFN (Figura 4). Estos resultados confirman los datos de expresión diferencial en arrays de expresión, e indican un papel de SIDT1 en la regulación de la respuesta a la estimulación de TLR-7 y -9, actuando específicamente en la vía de producción de interferones. To corroborate the result of the expression array, ODN2006 and imiquimod stimulations were performed on Gen2.2 cells and their controls for 24 hours, and several cytokines and chemokines were measured in the supernatant by ELISA assays (eBioscience), following the recommended protocol by the manufacturer. As illustrated in Figure 3, the stimulation of the production of TNFa, IL-6 and IL-8 proinflammatory cytokines is not altered by the silencing of SIDT1. However, there is a dramatic decrease in interferon type I (IFNal) and III (IFN 1), as well as CCL3 chemokine, which is regulated by IFN (Figure 4). These results confirm the differential expression data in expression arrays, and indicate a role of SIDT1 in regulating the response to stimulation of TLR-7 and -9, acting specifically in the interferon production pathway.
Inhibición parcial de la entrada de ODN2006 en células Gen2.2 shSIDTI. Dado que otros autores han emitido la hipótesis de que SIDT1 sería un canal de internalización de ácidos nucleicos, realizamos estudios de internalización mediante citometría de flujo. Las células Gen2.2 shSIDTI y sus controles se incubaron con el ligando de TLR-9 ODN2006 marcado con la molécula fluorescente Cy5, y la entrada se monitorizó mediante citometría de flujo en un citómetro FACS Canto (BD Biosciences). Como se muestra en la figura 5, el silenciamiento de SIDT1 induce una disminución de la entrada de ODN2006-Cy5 de un 30% aproximadamente, lo que no es suficiente para justificar la drástica regulación de IFN de tipo I y III, con conservación de la inducción de citoquinas proinflamatorias. Este resultado sugiere que SIDT1 podría estar influenciando la respuesta de IFN por un mecanismo diferente al transporte de ácidos nucleicos. El silenciamiento de SIDT1 bloquea la producción de IFN de tipo I en pDC primarias ex vivo. Partial inhibition of ODN2006 entry in Gen2.2 shSIDTI cells. Since other authors have hypothesized that SIDT1 would be a nucleic acid internalization channel, we conducted internalization studies using flow cytometry. Gen2.2 shSIDTI cells and their controls were incubated with the TLR-9 ODN2006 ligand labeled with the Cy5 fluorescent molecule, and the input was monitored by flow cytometry on a Canto FACS cytometer (BD Biosciences). As shown in Figure 5, the silencing of SIDT1 induces a decrease in the ODN2006-Cy5 input of approximately 30%, which is not sufficient to justify the drastic regulation of type I and III IFN, with conservation of the induction of proinflammatory cytokines. This result suggests that SIDT1 could be influencing the IFN response by a different mechanism to nucleic acid transport. SIDT1 silencing blocks the production of type I IFN in ex vivo primary pDCs.
Se aislaron pDC circulantes a partir de unidades de sangre de donantes sanos utilizando el BDCA-4 Positive Selection Kit en un separador Automacs Pro (ambos de Miltenyi Biotec), siguiendo las instrucciones del fabricante. En todos los casos se obtuvieron pDC con más de un 90% de pureza. Las pDC purificadas se trasdujeron con los lentivirus shSIDTI o shCTL a una moi de 0.4 y se estimularon con 1 M ODN2006 en medio RPMI suplementado con 5% de plasma autólogo inactivado durante 24h. La expresión de SIDT1 se cuantificó utilizando el Quantigene Assay Kit (Panomics/Affimetrix) y el luminómetro Glomax Multidetector System (Promega), siguiendo las instrucciones de los fabricantes. En esas condiciones se observó una disminución de la expresión de SIDT1 en las células silenciadas del 50% (Figura 6A). El IFN de tipo I del sobrenadante se cuantificó utilizando las células indicadoras de IFNa/β HEK-Blue IFNa/β (Invivogen), siguiendo las recomendaciones del fabricante. Como se observa en la figura 6B, el silenciamiento de SIDT1 en las pDC primarias induce una disminución de la producción de IFNa/β de tres órdenes de magnitud. Circulating pDCs were isolated from blood units from healthy donors using the BDCA-4 Positive Selection Kit in an Automacs Pro separator (both from Miltenyi Biotec), following the manufacturer's instructions. In all cases, pDCs with more than 90% purity were obtained. The purified pDCs were transduced with the shSIDTI or shCTL lentiviruses at a moi of 0.4 and were stimulated with 1 M ODN2006 in RPMI medium supplemented with 5% autologous inactivated plasma for 24 h. SIDT1 expression was quantified using the Quantigene Assay Kit (Panomics / Affimetrix) and the Glomax Multidetector System (Promega) luminometer, following the manufacturers instructions. Under these conditions, a decrease in SIDT1 expression was observed in the silenced cells of 50% (Figure 6A). Type I IFN of the supernatant was quantified using IFNa / β HEK-Blue IFNa / β (Invivogen) indicating cells, following the manufacturer's recommendations. As seen in Figure 6B, the silencing of SIDT1 in the primary pDC induces a decrease in IFNa / β production of three orders of magnitude.
Estos resultados utilizando pDC primarias ex vivo confirman los obtenidos con la línea célular modelo y demuestran que SIDT1 es un mediador fundamental para la inducción de una respuesta de interferón tras la estimulación de los receptores endosomales de ácidos nucleicos TLR-7 y -9. Determinación del papel de SIDT1 en los receptores endosomales de ácidos nucleicos y en la respuesta de IFN mediada por receptores citosólicos These results using ex vivo primary pDC confirm those obtained with the model cell line and demonstrate that SIDT1 is a fundamental mediator for the induction of an interferon response after stimulation of the endosomal receptors of TLR-7 and -9 nucleic acids. Determination of the role of SIDT1 in endosomal nucleic acid receptors and in the IFN response mediated by cytosolic receptors
Para determinar si el papel de SIDT1 está limitado a los receptores endosomales de ácidos nucleicos, o por el contrario está implicado también en la respuesta de IFN mediada por receptores citosólicos, se realizaron estimulaciones con ODN2006 y poli l:C encapsulados en liposomas mediante el uso de lipofectamina, para permitir el acceso de los ligandos al citosol. Como se muestra en la figura 7, las células Gen2.2 silenciadas para SIDT1 también mostraron una respuesta alterada tras estimulación de receptores citosólicos de DNA con CpG+lipofectamina. Sin embargo, la respuesta de IFN mediada por receptores citosólicos de RNA (poli l:C + lipofectamina) no se alteró en las células shSIDTI , que SIDT1 no es un mediador general de la respuesta de IFN frente a ácidos nucleicos exógenos, sino que es un mediador selectivo de ciertas vías de reconocimiento en las pDC. To determine whether the role of SIDT1 is limited to endosomal nucleic acid receptors, or on the contrary is also involved in the IFN response mediated by cytosolic receptors, stimulations with ODN2006 and poly l: C encapsulated in liposomes were performed through use. of lipofectamine, to allow the access of the ligands to the cytosol. As shown in Figure 7, Gen2.2 cells silenced for SIDT1 also showed an altered response after stimulation of cytosolic DNA receptors with CpG + lipofectamine. However, the IFN response mediated by cytosolic RNA receptors (poly 1: C + lipofectamine) was not altered in shSIDTI cells, which SIDT1 is not a general mediator of the IFN response to exogenous nucleic acids, but is a selective mediator of certain recognition pathways in the pDC.
Se han identificado varias moléculas clave en la trasducción de señales que dan lugar a la inducción de una respuesta de IFN tras la estimulación de TLR9. Dos de ellas fueron seleccionadas para el estudio del papel de SIDT1 mediante estudios de Western blot: TBK-1 activa la fosforilación de diferentes miembros de la familia IRF, permitiendo su traslocación al núcleo para activar los genes de la firma de IFN, y por tanto es fundamental para el desarrollo de una respuesta completa mediada por TLR9. Por otro lado, se ha demostrado que el factor de iniciación de la traducción de mRNA elF2a está implicado en la respuesta a la estimulación de TLR9 y TLR3. Como se refleja en la figura 8, la estimulación con ODN2006 induce la fosforilación de TBK-1 y elF2a. Sin embargo, las células shSIDI la fosforilación de las moléculas TBK1 y elF2a es deficiente. Estos resultados sugieren que SIDT1 es fundamental para una el establecimiento de una cascada de trasducción de señales óptima para la inducción de interferones de tipo I y III tras la estimulación de TLR7, TLR9 y receptores citosólicos de DNA. Several key molecules have been identified in signal transduction that lead to the induction of an IFN response after stimulation of TLR9. Two of them were selected for the study of the role of SIDT1 through Western blot studies: TBK-1 activates the phosphorylation of different members of the IRF family, allowing its translocation to the nucleus to activate IFN signature genes, and therefore It is essential for the development of a complete response mediated by TLR9. On the other hand, it has been shown that the translation initiation factor of mRNA elF2a is involved in the response to stimulation of TLR9 and TLR3. As reflected in Figure 8, stimulation with ODN2006 induces phosphorylation of TBK-1 and elF2a. However, shSIDI cells phosphorylation of TBK1 and elF2a molecules is poor. These results suggest that SIDT1 is essential for the establishment of an optimal signal transduction cascade for the induction of type I and III interferons after stimulation of TLR7, TLR9 and cytosolic DNA receptors.

Claims

REIVINDICACIONES
1.- El uso de una composición que comprende un agente modulador de la actividad de SIDT1 en la elaboración de un medicamento para el tratamiento, mejora o prevención de enfermedades infecciosas, inflamatorias o autoinmunes mediadas por IFN. 1.- The use of a composition comprising an agent modulating the activity of SIDT1 in the preparation of a medicament for the treatment, improvement or prevention of infectious, inflammatory or autoimmune diseases mediated by IFN.
2. - El uso de una composición según la reivindicación 1 , donde los agentes moduladores se seleccionan de la lista que consiste en: a) una molécula pequeña, b) una molécula de RNA, c) un oligonucleótido antisentido, d) un anticuerpo, e) una ribozima, f) una endonucleasa capaz de realizar edición génica, o cualquiera de sus combinaciones. 2. - The use of a composition according to claim 1, wherein the modulating agents are selected from the list consisting of: a) a small molecule, b) an RNA molecule, c) an antisense oligonucleotide, d) an antibody, e) a ribozyme, f) an endonuclease capable of gene editing, or any combination thereof.
3. - El uso de una composición según cualquiera de las reivindicaciones 1 -2, donde los agentes moduladores se seleccionan de la lista que consiste en: SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID3. - The use of a composition according to any of claims 1-2, wherein the modulating agents are selected from the list consisting of: SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID
NO: 5, o cualquiera de sus combinaciones. NO: 5, or any of its combinations.
4. - El uso de una composición según cualquiera de las reivindicaciones 1 -3, donde la enfermedad inflamatoria o autoinmune mediada por IFN se selecciona de la lista que comprende: alopecia areata, la espondilitis anquilosante, síndrome antifosfolípidos, enfermedad autoinmune de Addison, enfermedades autoinmunes de la glándula suprarrenal, la anemia hemolítica autoinmune, hepatitis autoinmune, ooforitis autoinmune y orquitis, trombocitopenia autoinmune, enfermedad de Behcet, penfigoide bulloso, cardiomiopatía, enfermedad celíaca , la dermatitis , la fatiga crónica , síndrome de disfunción inmune ( SFC ), la polineuropatía desmielinizante inflamatoria crónica , síndrome de Churg -Strauss, penfigoide cicatricial, síndrome de CREST, enfermedad por crioaglutininas, lupus discoide, crioglobulinemia mixta esencial, fibromialgia-fibromiositis, glomerulonefritis, enfermedad de Graves, el síndrome de Guillain-Barre, tiroiditis de Hashimoto, fibrosis pulmonar idiopática, púrpura trombocitopénica idiopática (PTI), la neuropatía por IgA, artritis juvenil, artritis reumatoide, el liquen plano, enfermedad de Meniere, enfermedad mixta del tejido conectivo, esclerosis múltiple , diabetes mellitus inmunomediada o tipo 1 , miastenia gravis, pénfigo vulgar, la anemia perniciosa, la poliarteritis nodosa, policondritis, síndromes poliglandulares, polimialgia reumática, la polimiositis y la dermatomiositis, agammaglobulinemia primaria, cirrosis biliar primaria, psoriasis, artritis psoriásica, el fenómeno de Raynauld , síndrome de Reiter, la sarcoidosis, la esclerodermia, la esclerosis sistémica progresiva, síndrome de Sjógren, síndrome de Goodpasture , el síndrome del hombre rígido, lupus eritematoso sistémico, lupus eritematoso, la arteritis de Takayasu, arteristis temporal/arteritis de células gigantes , colitis ulcerosa , uveítis, vasculitis como la vasculitis dermatitis herpetiforme, vitíligo , granulomatosis de Wegener, Enfermedad de la Membrana basal glomerular, síndrome antifosfolípido, Enfermedades Autoinmunes del Sistema Nervioso, fiebre mediterránea familiar, el síndrome miasténico de Lambert- Eaton, la oftalmía simpática , poliendocrinopatías , psoriasis, o cualquiera de sus combinaciones. 4. - The use of a composition according to any of claims 1 -3, wherein the IFN-mediated inflammatory or autoimmune disease is selected from the list comprising: alopecia areata, ankylosing spondylitis, antiphospholipid syndrome, Addison autoimmune disease, diseases Autoimmune of the adrenal gland, autoimmune hemolytic anemia, autoimmune hepatitis, autoimmune oophoritis and orchitis, autoimmune thrombocytopenia, Behcet's disease, bullous pemphigoid, cardiomyopathy, celiac disease, dermatitis, chronic fatigue, immune dysfunction syndrome (CFS), Chronic inflammatory demyelinating polyneuropathy, Churg-Strauss syndrome, scarring pemphigoid, CREST syndrome, cryogglutinin disease, discoid lupus, essential mixed cryoglobulinemia, fibromyalgia-fibromiositis, glomerulonephritis, Graves disease, Guillain-Barreim syndrome, thyroiditis, thyroiditis idiopathic pulmonary fibrosis, purpur to idiopathic thrombocytopenic (PTI), IgA neuropathy, juvenile arthritis, rheumatoid arthritis, lichen planus, Meniere's disease, mixed connective tissue disease, multiple sclerosis, immunomediated or type 1 diabetes, myasthenia gravis, pemphigus vulgaris, anemia pernicious, polyarteritis nodosa, polychondritis, polyglandular syndromes, polymyalgia rheumatica, polymyositis and dermatomyositis, primary agammaglobulinemia, Primary biliary cirrhosis, psoriasis, psoriatic arthritis, Raynauld's phenomenon, Reiter's syndrome, sarcoidosis, scleroderma, progressive systemic sclerosis, Sjógren's syndrome, Goodpasture's syndrome, rigid man's syndrome, systemic lupus erythematosus, lupus erythematosus, Takayasu arteritis, temporal arteristis / giant cell arteritis, ulcerative colitis, uveitis, vasculitis such as vasculitis, herpetiform dermatitis, vitiligo, Wegener granulomatosis, Glomerular basement membrane disease, antiphospholipid syndrome, Autoimmune diseases of the Nervous System, Mediterranean fever , the Lambert-Eaton myasthenic syndrome, sympathetic ophthalmia, polyocrinopathies, psoriasis, or any combination thereof.
5.- El uso de una composición según cualquiera de las reivindicaciones 1 -4, donde la enfermedad inflamatoria o autoinmune mediada por IFN es el lupus eritematoso sistémico (LES). 5. The use of a composition according to any of claims 1-4, wherein the inflammatory or autoimmune disease mediated by IFN is systemic lupus erythematosus (SLE).
6.- El uso de una composición según cualquiera de las reivindicaciones 1 -4, donde la enfermedad inflamatoria o autoinmune mediada por IFN es la psoriasis. 6. The use of a composition according to any of claims 1-4, wherein the inflammatory or autoimmune disease mediated by IFN is psoriasis.
7.- Un método de selección de agentes terapéuticos útiles en el tratamiento de enfermedades infecciosas, inflamatorias o autoinmunes mediada por IFN, que comprende: a) poner en contacto el compuesto a analizar con el polipéptido SIDT1 , y b) detectar la unión de dicho compuesto a analizar con el polipéptido SIDT1. 7. A method of selecting therapeutic agents useful in the treatment of infectious, inflammatory or autoimmune diseases mediated by IFN, comprising: a) contacting the compound to be analyzed with the SIDT1 polypeptide, and b) detecting the binding of said compound to be analyzed with the SIDT1 polypeptide.
8. Un método de selección de agentes terapéuticos útiles en el tratamiento de enfermedades infecciosas, inflamatorias o autoinmunes mediada por IFN, que comprende: a) determinar la actividad de SIDT1 a una determinada concentración del compuesto a analizar o en ausencia de dicho compuesto, b) determinar la actividad de SIDT1 a una concentración del compuesto a analizar diferente de la de a). 8. A method of selecting therapeutic agents useful in the treatment of infectious, inflammatory or autoimmune diseases mediated by IFN, comprising: a) determining the activity of SIDT1 at a certain concentration of the compound to be analyzed or in the absence of said compound, b ) determine the activity of SIDT1 at a concentration of the compound to be analyzed different from that of a).
9. Un método de selección de agentes terapéuticos útiles en el tratamiento de enfermedades virales, inflamatorias o autoinmunes mediada por IFN, que comprende: a) determinar la actividad de SIDT1 a una determinada concentración del compuesto a analizar, b) determinar la actividad de SIDT1 en presencia de un compuesto que se conoce que modula la actividad de SIDT1. 9. A method of selecting therapeutic agents useful in the treatment of viral, inflammatory or autoimmune diseases mediated by IFN, comprising: a) determining the activity of SIDT1 at a certain concentration of the compound to be analyzed, b) determining the activity of SIDT1 in the presence of a compound that is known to modulate the activity of SIDT1.
10. Un método de selección de agentes terapéuticos útiles en el tratamiento de enfermedades infecciosas, inflamatorias o autoinmunes mediada por IFN, que comprende: a) poner en contacto el compuesto a analizar con el polinucleótido SIDT1 , b) detectar la unión de dicho compuesto a analizar con el polinucleótido SIDT1. 10. A method of selecting therapeutic agents useful in the treatment of infectious, inflammatory or autoimmune diseases mediated by IFN, comprising: a) contacting the compound to be analyzed with the SIDT1 polynucleotide, b) detecting the binding of said compound to be analyzed with the SIDT1 polynucleotide.
1 1. Un método para la obtención de datos útiles en el diagnóstico y/o pronóstico y/o de respuesta al tratamiento de enfermedades virales, inflamatorias o autoinmunes mediada por IFN, que comprende: a) determinar la expresión de SIDT1 en una muestra extraída de un mamífero, b) comparar los valores de la expresión de SIDT1 obtenidos en a) con los valores estándar en mamíferos sanos o enfermos. 1 1. A method for obtaining useful data in the diagnosis and / or prognosis and / or response to the treatment of viral, inflammatory or autoimmune diseases mediated by IFN, comprising: a) determining the expression of SIDT1 in an extracted sample of a mammal, b) compare the values of SIDT1 expression obtained in a) with the standard values in healthy or diseased mammals.
PCT/ES2015/070369 2014-05-07 2015-05-07 Composition for modulating the activity of sidt1 for the treatment of ifn-mediated infectious, inflammatory or autoimmune diseases WO2015169994A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
ES201430669 2014-05-07
ESP201430669 2014-05-07

Publications (1)

Publication Number Publication Date
WO2015169994A1 true WO2015169994A1 (en) 2015-11-12

Family

ID=54392189

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/ES2015/070369 WO2015169994A1 (en) 2014-05-07 2015-05-07 Composition for modulating the activity of sidt1 for the treatment of ifn-mediated infectious, inflammatory or autoimmune diseases

Country Status (1)

Country Link
WO (1) WO2015169994A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115697953A (en) * 2020-06-08 2023-02-03 公立大学法人富山县立大学 Novel compounds as inhibitors of Toll-like receptor 7 activation

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010057217A1 (en) * 2008-11-17 2010-05-20 Alnylam Pharmaceuticals, Inc. Compositions and methods for increasing cellular uptake of rnai via sid-1
US20100152280A1 (en) * 2004-05-24 2010-06-17 Isis Pharmaceuticals, Inc. Modulation of sid-1 expression

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100152280A1 (en) * 2004-05-24 2010-06-17 Isis Pharmaceuticals, Inc. Modulation of sid-1 expression
WO2010057217A1 (en) * 2008-11-17 2010-05-20 Alnylam Pharmaceuticals, Inc. Compositions and methods for increasing cellular uptake of rnai via sid-1

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
DUXBURY M S ET AL.: "RNA interference: A mammalian SID-1 homologue enhances siRNA uptake and gene silencing efficacy in human cells.", BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS, vol. 331, no. 2, 3 June 2005 (2005-06-03), ORLANDO, FL, US, pages 459 - 463, XP004867577, ISSN: 0006-291X *
HORNUNG V ET AL.: "Sequence-specific potent induction of IFN-alpha by short interfering RNA in plasmacytoid dendritic cells through TLR7.", NATURE MEDICINE, vol. 11, no. 3, 20 February 2005 (2005-02-20), NEW YORK, NY, US, pages 263 - 270, XP002505448, ISSN: 1078-8956 *
MARY K CROW ET AL.: "Interferon-alpha: a therapeutic target in systemic Lupus erythematosus.", RHEUMATIC DISEASES CLINICS OF NORTH AMERICA, vol. 36, no. 1, 1 February 2010 (2010-02-01), PHILADELPHIA, PA, US, pages 173 - 186, XP002661031, ISSN: 0889-857X *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115697953A (en) * 2020-06-08 2023-02-03 公立大学法人富山县立大学 Novel compounds as inhibitors of Toll-like receptor 7 activation

Similar Documents

Publication Publication Date Title
US10184151B2 (en) Micrornas in neurodegenerative disorders
WO2017191274A2 (en) Rna encoding a therapeutic protein
US9828601B2 (en) Compositions for inhibiting checkpoint gene expression and uses thereof
US7928083B2 (en) H19 silencing nucleic acid agents for treating rheumatoid arthritis
JP2021167353A (en) Compositions and methods for treating autoimmune diseases and cancers
CN107532169B (en) Dynamin 2 inhibitors for the treatment of duchenne&#39;s muscular dystrophy
US20190300882A1 (en) Methods of treating neuroblastoma and reagents therefor
WO2017005773A1 (en) Use of catenin- beta 1-targeting micrornas for treating liver cancer
WO2017205765A1 (en) Compositions and methods for inhibiting ptpn22
JP2021505200A (en) Compositions and Methods for Treating Genomic Imprinting Disorders
US20190055556A1 (en) Targeting pleiotrophin signaling to limit high-grade glioma invasion
JP6975465B2 (en) Short hairpin RNA (SHRNA734) and its use to positively select and eliminate genetically modified cells
KR20210123338A (en) Calcineurin Inhibitor Resistant Immune Cells for Use in Adoptive Cell Transfer Therapy
US20210369761A1 (en) Methods of treating motor neuron diseases
KR102353847B1 (en) Reagents for the treatment of oropharyngeal muscular dystrophy (OPMD) and uses thereof
KR20230004456A (en) Methods and compositions for sensitization of tumor cells to immunotherapy
US20140093494A1 (en) Alpha synuclein toxicity
US20210380978A1 (en) The long non-coding RNA INCA1 and Homo sapiens heterogeneous nuclear ribonucleoprotein H1 (HNRNPH1) as therapeutic targets for immunotherapy
WO2015169994A1 (en) Composition for modulating the activity of sidt1 for the treatment of ifn-mediated infectious, inflammatory or autoimmune diseases
WO2020128006A1 (en) Mirna for use in therapy
WO2010034779A2 (en) Composition and method for treatment of preterm labor
EP3999098A1 (en) Methods of treating pain
US20190263906A1 (en) Immune modulators for reducing immune-resistance in melanoma and other proliferative diseases
US11273176B2 (en) Use of PLA2G5-deficient suppressive macrophages in suppression of inflammation
WO2024059821A2 (en) Car t cell compositions for treatment of cancer

Legal Events

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

Ref document number: 15788915

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

32PN Ep: public notification in the ep bulletin as address of the adressee cannot be established

Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 112(1) EPC (EPO FORM 1205A DATED 10.03.2017)

122 Ep: pct application non-entry in european phase

Ref document number: 15788915

Country of ref document: EP

Kind code of ref document: A1